HEMA1 2 NOTES Compressed 101 321

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 221

- Formed w/in the macrophages & reduced into - epsilon chain = counterpart of the gamma, beta

bilirubin and delta chains

2 TYPES OF BILIRUBIN 3 EMBRYONIC HEMOGLIBIN:

1. Conjugated - DIFFERENCE: Composition of globin structure


2. Unconjugated a. Hb Gower I (2 zeta & 2 epsilon)
- What comes out of bilirubin b. Hb Gower II (2 alpha & 2 epsilon)
- Non-polar water insoluble & when numerous in c. Hb Portland (2 zeta & 2 gamma)
plasma it causes jaundice 2. Hb F (2 alpha & 2 gamma)
- Major Hb of the fetus and the newborn
a. Unconjugated bilirubin will be released in the - Has high affinity to O2 thus it will not easily
plasma & for it to be normally excreted it will be release the O2
brought by albumin (transport protein) into the - Resistant to both acid elution and alkali
liver for conjugation denaturation
b. In the liver w/ UDP-glucorynyl transferase which - After birth, smaller amounts of HbF are
will catalyze the addition of 2 glucoronic acids to produced
bilirubin forming now the conjugated - Diseases associated: HPFH & B. Thalassemia
bilirubin/bilirubin diglucuronide which is water - Increased would lead to hypoxia
soluble - In fetus, anaerobic respiration occurs, for it to
c. Bilirubin diglucuronide will be stored in the bile. conserve its O2, it contains Hb F, but for new
COMPOSITION OF BILE: bile salts, bile acids, bile born, aerobic respiration, Hb F is still increased
pigments (conjugated bilirubin) thus decreased O2, this decrease is
FUNCTION OF BILE: digestion of fats compensated by the increased Hb F
d. During digestion, bile is released into the small
AFTER BIRTH, SMALLER AMOUNTS OF HbF ARE
intestines (duodenum) via the common bile
PRODUCED:
duct.
e. Bilirubin will be acted by bacterial intestinal o 6 months = <8%
enzymes converting it into urobilinogen. ½ of o After age 2 years = <2%
urobilinogen will be reabsorb to the liver for o Adults = < 1-2%
reprocessing while some will be filtered by the
kidneys and be excreted into the urine GLOBIN SYNTHESIS:
f. Urine urobilinogen will give off the dark yellow - Hb F, Gower 1 & 2 & Portland are Hb that are
color of urine (normal) synthesized separately & produced separately
- Primitive RBC are the very first blood cell
g. The other half will be excreted into the feces
produced during the mesoblastic stage which
giving a color of light yellow, light brown to dark
will oxygenate the embryo’s tissue. This
brown due to urobilin and stercobilin coming primitive RBC contains the embryonic Hb
from urobilinogen
a. Zeta & Epsilon are synthesized in the
INTRAVASCULAR HUMAN HEMOGLOBIN
mesoblastic stage forming the Gower 1
- Same w/ extravascular but w/out the
Note: synthesis of zeta & epsilon is high at the
macrophages. start but declines thereafter, thus decreasing
- Lysis occurs in the circulation embryonic Hb.
NORMAL HUMAN HEMOGLOBINS: b. After few days, alpha chains are synthesized
wherein a small amount of it combines w/
1. EMBRYONIC HEMOGLOBIN epsilon forming the Gower 2
- Produced in the mesoblastic stage c. A small amount of Gamma is synthesized &
- First 3 months after conception combines w/ zeta forming the Portland
- zeta chain = analogue of a-chain d. No synthesis of Epsilon and Zeta would lead to
dissolution of embryonic hb wherein it only
persists for at least 3 months. As a  Methemoglobin is not a compound but only
counterpart, the A & Gamma increases. an Hb containing an oxidized Fe (Ferric)
e. Combination of these 2 forms the Hb F during  Sulfhemoglobin is a Hb w/ sulfide radical
the hepatic stage  Oxy & DeoxyHb are normal Hb formed during
f. Synthesis of A chain persist until after birth the normal transport of gases. These are
while gamma starts to decrease after birth & normal both in presence & function.
as a counterpart B chain gradually increases.  Carboxy, Met & SulfHb are abnormal in
g. Combination of A & B chain forms the Hb A1. function but normal in presence. These are
h. Delta chain, does not peak & combination of present in small amount in circulation.
this to A chain will form the Hb A2  Affinity of carboxyhemoglobin to Hb is >200x
thus instead that O2 binds,
carboxyhemoglobin binds w/ Hb thus it has an
abnormal function & is present in very minute
3. Hb A1 (2A & 2B): Major normal adult hemoglobin
concentration.
(97%)
 Everyday 3.5% of Hb is converted into
4. Hb A2 (2A & 2D): methemoglobin but since RBC have reducing
o Accounts for 1.5 % - 3.5 % of normal capacity, it is reduced back into a normal Hb.
adult Hb Deficiency in reducing capacity of RBC, RBC
o Increased in some – B Thalassemia’s, can no longer reverse the reaction thus
hyperthyroidism and in some cases of increase in methemoglobin causing
megaloblastic anemia methemoglobinemia.
 Affinity of Hb to O2 is dependent in the state
HEMOGLOBIN MOLECULAR STAGE OF LIFE of Fe – ferrous
STRUCTURE  Oxidation reduction is reversible, meaning it is
oxidized but it is also reduced.
Gower 1 2Z&2E EMBRYONIC
 Sulfhemoglobins presence & function is both
Gower 2 2A&2E EMBRYONIC abnormal thus it must not be present in
circulation. It is the only Hb that is not
Portland 2Z&2G EMBRYONIC measured by cyanmethemoglobin procedure.
 Conversion of Hb to Sulf is irreversible.
Fetal (F) 2A&G FETAL

A1 2A&2B ADULT
FORMS OF HEMOGLOBIN
A2 2A&2D ADULT NORMAL

1. OXYHEMOGLOBIN (HbO2)
- Circulates in the arterial circulation
HEMOGLOBIN COMPOUNDS - Formed when the Hb passes through the
OXYHEMOGLOBIN O2 + Hb Hb F, A1 & pulmonary capillaries of the lungs
- The O2 is loosely bound and unstable for easier
A2
release into the tissues
DEOXYHEMOGLOBIN CO2 + Hb Hb F, A1 & - Gives a scarlet red/bright red color
A2
NOTE:
CARBOCYHEMOGLOBIN CO + Hb Hb A2 Basilic is not appropriate for blood collection due to
possibility that brachial artery might be hit
METHEMOGLOBIN Ferric Hb Hb A1 & A2

SULFHEMOGLOBIN S + hb Hb A1 & A2 2. DEOXYHEMOGLOBIN


- Reduced form however increased CO2
- Hemoglobin with CO2 (carbaminohemoglobin)
NOTE: wherein CO2 binds to the free amino group of
the Hb to form carbaminohemoglobin
- Gives a dark red color due to presence in venous NOTE: RAPID TESTS FOR HbCO
circulation a. NaOH test
- Formed when Hb passes through the tissues - Composition: 40% NaOh + EDTA – blood
- Process: warm gently
ABNORMAL - Result: HbCO: red color HbO2: Black-Brown
1. CARBOXYHEMOGLOBIN (HbCO2)
b. Dilution test (addition of water)
- Hemoglobin with CO (affinity of CO to Hb is 200-
- Compostion: 1 mL blood + 50 mL water
210x greater than the affinity of O2 thus it binds
- Result:
more in Hb) o HbCO: cherry red – pink or bluish red
- Normally present in body due to endogenous CO
source (<1%) but have an abnormal function o HbO2: Yellowish Red
because it cannot bind with O2
- From the breakdown of protoporphyrin into c. Tannic Acid test
biliverdin & CO which is the endogenous source - Composition: 1% Tannic acid which will tend
which is only minimal (<1%). At the same time, to acid digest the blood resulting to black-
for people who are exposed to normal brown color
atmospheric O2, the half-life of CO is only 4 - Result:
hours & 50% is lost by exhalation & this is not o HbCO: red precipitate
enough to cause CO poisoning o HbO2: Black-brown discoloration
2. METHEMOGLOBIN/ HEMIGLOBIN (Hi)
- Chief Sources of CO (exogenous): automobile
- Iron (Fe++) is oxidized into its Fe+++ form
exhaust, industrial wastes, tobacco smoking
- Not a compound but an oxidized Hb
(10% of Hb of people expose to exogenous
- Absorption wavelength: 630 – 635 nm
source)
- Screening tests:
- Imparts a cherry red in color in blood & skin
o Aeration: blood retains its color
- Absorption wavelength: 576 nm
o Addition of 1% MB: blood turns red
CRITICAL VALUE: - Have an abnormal function but normally present
5g /100 ml which may cause irreversible tissue in blood w/ conc. of 1.5% because every day
changes since COHb cannot bind w/ O2, thus it cannot almost 0.5 – 3% Hb is converted to hemoglobin
transport O2 thus it cannot oxygenate the tissues & that does not accumulate even if it is oxidized it
lungs. There are body tissues that are very dependent is reduced back to normal Hb as oxidation
on O2 (brain cells). When brain cells are depleted w/ reduction is reversible. But if amount of
O2 this will result to cell damage & brain cell damage oxidation is >1.5-3.5% g/dL (10%) it will cause
is irreversible. If values are >50 – 70% this may lead to
cellular damage causing cyanosis wherein the
asphyxiation which is experienced by a person being
patient will suffer from bluish discoloration of
strangled, it is a condition wherein the patient is
depleted w/ O2 thus the patient will lose the skin as well as difficulty of breathing due to
consciousness & may lead to death, however depletion of O2.
treatment may be employed w/ hyperbaric O2 - If present in blood at high conc. >1.5-3.5% g/dL
(10%) this will result to a chocolate brown color
Treatment: administration of hyperbaric O2 – involves
MEDICAL IMPORTANCE:
high amount of conc. O2 w/ pressure in a pressurize
room in a pressurize tube. Although the affinity of - With strong affinity w/ cyanide forming
COHb is greater than O2, w/ higher amount of hemoglobincyanide (HiCN) which may take
pressurize O2, the O2 exposure can displace CO from place inside the body. It is important in the
attachment to Hb thus bringing the CO to its normal treatment of cyanide poisoning. Free cyanide
Hb range in the circulation is very toxic.
MANAGEMENT OF CYANIDE POISONING
- NO2 is given which will cause the oxidation.
NO2 will convert Hb into hemiglobin causing
oxidation of the patients Hb thus once
hemiglobin is formed, it can bind to the OTHER REDUCING AGENTS:
produced cyanide forming into HiCN. The free a. METHYLENE BLUE
poisonous cyanide is converted into less - Often given to patients suffering from
poisonous cyanide. methemoglobinemia
- It will cause the reaction of glucose 6-P to be
faster wherein the H of NADH will be
 Maintenance of Hemoglobin in the Normal transferred to the Hb thus reducing Hb
Reduced state (Fe++): Reducing properties of however the transfer of H is slow wherein
RBC methylene blue will enhance this reaction
b. ASCORBIC ACID: a potent reducing agent &
PRODUCTION OF:
also an anti-oxidant
 Reduced Diphosphopyridine dinucleotide
(DPNH) in the presence of methemoglobin
METHEMOLOBINEMIA
reductase (diaphorase)
- Reaction in Embden-Meyerhof Pathway that - increase in the methemoglobin conc. in blood
Generates NADH:
CONDITIONS OF METHEMOGLOBINEMIA
o NAD is reduced into NADH which has a
reducing potential thus deficiency of it a. Inherited (enzyme deficiency): commonly
will not make it to reduce the ferric into attributed to NADH-Methemoglobin reductase
ferrous. deficiency/Diaphorase deficiency which is
important in reducing ferric to ferrous
 Reduced Triphosphopyridine nucleotide (TPNH) b. Inherited M (methemoglobin)
in the presence of Glucose-6-PO4 - results of various amino acid substitutions in the
dehydrogenase globin chain that directly affect the heme group
- produced in Embden-Meyerhof Pathway in the - due to increase methemoglobin, RBC cannot
Hexose Monophosphate shunt keep up thus it cannot reduced back to
- Process: from phosphorylation of glucose to hemoglobin
glucose 6 phosphate it enters the hexose
METHEMOGLOBIN VARIANTS
monophosphate shunt & converted into 6-P
gluconate catalyze by G-6-PD with a coenzyme - Different structural abnormality but are not
NADP wherein in this reaction it is reduced which protected from being oxidized meaning they are
is similar to TPNH. With the reduced NADP it can easily oxidized
caused the production or reduction of more 1. M-saskatoon
glutathione which is also a reducing potential. 2. M-boston
The major reducing capacity of RBC is found in 3. M-iwate
the hexose monophosphate shunt because in 4. M-hydePark
this shunt, it can be able to produce 1.) NADPH 5. M-milwaekee
& 2.) GSH
c. Acquired:
 Glutathione and its associated enzymes - effects of chemical or therapeutic agents (aniline
o Glutathione is the major anti-oxidant dyes in food, nitrate & nitrite-rich water and
since it has a reducing property w/ foodstuffs ; anti-malarial drugs & sulfonamides)
NADPH - Therapy: Ascorbic acid & Methylthionimium Cl
o Reduced glutathione is important in
protecting the Hb from being oxidized NOTE:
 Methylene blue is not a reducing agent but
NOTE: only accelerates the reaction.
Reduced NAD can bring back methemoglobin into  Carboxyhemoglobin and methemoglobin are
normal Hb, ferric to ferrous present in small quantities in the serum
however its function is abnormal.
SCREENING TEST FOR METHEMOGLOBIN 123 – 153  Higher in the morning & lower in
Composition: 1% Methylene Blue g/L the evening
Result: red or pink color of blood
AT BIRTH:  Lower if lying down

3. SULFHEMOGLOBIN (SHb) 150 – 200  Increased in smokers


- Formed when organic sulfides combine with Hb g/L
 Increased in high altitude
- Observed in patients taking in oxidant drugs
because the atmosphere is very
(phenacetin & acetanilid, sulfonamides)
thin thus O2 is thin
- Once Hb is converted into sulfhemoglobin it
remains as it is thus it is Irreversible for 120 days  Estrogen tends to inhibit
however it can combine w/ CO forming erythropoietin while androgen
carboxysulfhemoglobin will tend to stimulate more
- It is the irreversible formation which prevents it
to be measured by drabkins reagent thus it
cannot be converted in cyanmethemoglobin OTHER FORMS OF HEMOGLOBIN
- NOT normally present in the blood thus its a. CYANMETHEMOGLOBIN
presence at 0.5 g/100 mL is a critical level - Feriicyanide + Fe++ of Hb
causing damage to tissues - An in vitro test that may also occur inside the
- Imparts a greenish color body (in vivo)
- Unstable Hb that can easily precipitate into a - Its presence can also be induced in vivo (inside
Heinz bodies, an inclusion bodies, which when the body) especially during cyanide poisoning.
present to RBC, they tend to attach themselves - The most stable among the Hb pigments thus
on RBC membrane making RBC membrane rigid when performing cyanmethemoglobin
thus cells w/ Heinz bodies are prone to hemolysis procedure the Hb result can be delayed because
leading to hemolytic anemia. Even normal Hb the color produced does not fade
can form few Heinz bodies when denatured - Absorption wavelength: 540 nm
- Absorption wavelength: 600 - 620 nm

RFERENCE INCREASED Hb DECREASED Hb b. GLYCOSYLATED HEMOGLOBINS


VALUES - Also called “Glycated Hemoglobin” or simple
FOR  Polycythemi  All “Glycohemoglobin”.
HEMOGLO a - increase anemia - Described as “fast hemoglobins” because they
BIN RBC migrate fast in electrophoresis.
 Leukemi
production - Irreversibly glycosylated at 1 or both N-terminal
a
valines (or lysine) of the β-chains
 Dehydration
 Oligochr - Refers to the addition of a glucose molecule to
– due to
omia the hemoglobin molecule. Such addition is non-
hemoconcen
enzymatically catalyzed. The only basis of when
tration w/
the glucose binds in hemoglobin is the level of
decrease
plasma glucose that when the levels of plasma
plasma
glucose is elevated (like in hyperglycemia, DM
volume due
Type I and II), the excessive glucose in the plasma
to burns &
can bind with the hemoglobin called glycation or
diarrhea
glycosylation which is irreversible wherein once
MALE: PHYSIOLOGIC VARIATIONS the glucose is added to the Hgb molecule, this
Hgb will remain to be glycated so that the RBC,
140 – 175  After 50 years of age – slight once its Hgb is glycosylated, it will remain
g/L decrease glycolyated during the entire life span of the
FEMALE: RBC. This will now lead to the formation of:
o Hb A1a
o Hb A1b days. It is important index in diabetes control,
o Hb A1c: the FBS & RBS are more accurate as an index of
 Major fraction which serves as metabolic control for the past 2-4 months,
an index of the metabolic meaning the glycosylated Hb is the same for the
control over that last 2-4 next 4 months.
months. This is more preferred
OXYGEN DISSOCIATION CURVE
in monitoring glycemic control.
Oxyhemoglobin
 Elevated 2-3 folds in patients
dissociation curve
with Diabetes Mellitus (DM).
(ODC), is a curve that
 Preferred than FBS and RBS.
plots the proportion of
NOTE: HBA1c hemoglobin in its
saturated form on the
 Patients with DM are being monitored
Y axis against the
for their glycemic control and if their
prevailing oxygen
medications can control the increase
tension on the X axis. It
of their glucose. The monitoring
is slightly S shape.
should be a form of review on their
Left shift indicates increase O2 saturation
conditions for the past 2-4 months.
Right shift indicates that O2 is released in the tissues.
 The present HbA1c levels of the
This relates oxygen saturation and partial pressure of
patient reflects on his/her HbA1c
O2 in the blood (PO2), and is determined by
levels for the past 2-4 months.
hemoglobin affinity for oxygen wherein the higher the
 HbA1c is just for monitoring and not
affinity, the curved shift towards the left & vice versa.
for diagnosis for DM.
Affinity is the main factor that influences the shifting.

- Necessary for diabetic individual


FACTOR SHIFT CAUSED BY
- Fast Hb
- Hb w/ attached glucose which occurs when FACTOR FACTOR
plasma glucose is high. Glycation is not INCREASE DECREASE
enzymatically catalyzed. When glucose attached
to Hb, the only factor which will promote this is pH Left shift due to Right shift due
the increased in plasma glucose or increase affinity to decrease pH –
hyperglycemia. – alkalosis acidosis

NOTE: 2,3 DPH/BPG Right shift due do Left shift due to


 To which part of the Hgb will the glucose decrease affinity increase affinity
attach? due to increase due to decrease
o Glucose attaches at 1 or both N- DPG DPG
terminal valines (or lysine) or the β-
CO2 (haldane Right shift due to Right shift due
chains.
effect), H+ & increase of these to decrease of
 Only HbA1 are glycosylated Hgb because the
glycosylation occurs in the β-chain. Therefore, Cl- - all factors thus salt these factors
only Hgb has the β-chain. maintains the bridge is thus salt bridge
salt bridge stabilized thus is not stabilized
decreasing the thus increasing
2 REACTIONS ARE INVOLVED: affinity the affinity
1. REVERSIBLE Blood (Body) Right shift due to Left shift due to
2. AMADORI: irreversible meaning the glucose Temperature increase decreased
attached to the terminal valine or lysine can no Temperature temperature
longer be detached. & once the Hb is Rule: the
glycosylated it will remain glycosylated until 120 higher the
temperature,
the faster is DISORDERS RELATED WITH ABNORMAL
the HEMOGLOBINS
metabolism 1. THALASSEMIA
- Synthesis defect
Hb F Left shift due to Right shift due - Decreased or non-existent production of one or
increase Hb F to decreased Hb more globin chain type
-Has innate
thus it will not F thus it will - For both α & β, there is a deletion in the gene
increased
easily release O2 easily release resulting to incomplete synthesis of A & B chain.
affinity which
O2 - Diverse group of genetic disorders characterized
is needed by
fetus in an by a primary, quantitative reduction in globin
anaerobic chain synthesis for hemoglobin.
environment CLASSIFICATIONS:
in the uterus
thus if A. ALPHA THALASSEMIA
increased in - Indication of Deletion:
adults, this 1. Silent carrier - (_A/AA) (AA/_A) = ¼ thus no
may lead to signs & symptoms of anemia
deoxygenatio 2. Thalassemia trait/A-Thal minor – (_ _/AA)
n. (_A/A_) = 2/4 w/ mild signs & symptoms
3. Hb H disease – (_ _ /A _) = ¾ thus the
Abnormal Hbs production of Alpha chain becomes too
(depends on insufficient & B chain will be used in excess
characteristic resulting to Hb having all B chains (4 B)
of 4. Bart’s Hydrops fetalis – (_ _/ _ _) = 4/4 thus
abnormality) no synthesis of A chain, as early as fetal life,
Hb F which consist of 2 A & 2 G, there would
be no Hb F synthesis & what will be
SEVERAL ABNORMALITIES OF HEMOGLOBIN synthesize is the G chain (4 G).
1. HEME IS ABNORMAL
- Iron is deficient thus affecting Hb synthesis B. BETA THALASSEMIA
leading to IDA - Indication of Deletion: 0 superscript
- Deficient Protoporphyrin IX because of enzyme 1. Beta Thalassemia Major/Cooley’s anemia –
defect along the synthesis leading to excess iron B0/B
& iron will not be utilized in the absence of 2. Beta Thalassemia Intermedia – B+/B+,
protoporphyrin 9. Unused iron will precipitate present but cannot effect the synthesis of B
into siderotic granules leading to sideroblastic chain because of partial suppression.
anemia 3. Beta Thalassemia Minor/Cooley’s trait -
B0/B0, the patient is devoid of Hb A1
NOTE:
because the patient cannot synthesize B
Sideroblastic Anemia – problem in the utilization of
chain & what is elevated is Hb F.
iron due to deficiency in protoporphyrin 9

2. HEMOGLOBINOPATHIES
2. GLOBIN IS ABNORMAL - Disorder in structure
- Ex.: Hb A1 – 2 A w/ 4 genes & 141 amino acids & - Results from the alteration of the DNA genetic
2 B chains w/ 2 genes & 146 amino acids code for the chains
- 2 FACTORS: - Conditions characterized by qualitative
o Alpha Thalassemia structural abnormalities of the globin
o B Thalassemia polypeptide chains that result from alteration of
the DNA genetic code for those chains. Structural Hb S (SICKING HB)
abnormalities may involve amino acid. - The most well-known hb variant characterized
by substitution of glutamic acid by tRNA to valine
STRUCTURAL DEFECTS ON A OR B CHAIN RESULTING TO
on the 6th amino acid position of the β-chain.
Hb VARIANT:
Glutamic: CAG Hb S: β 6(A3) Glu → val
Valine: GUG
1. SUBSTITUTION
- Ex.: Hb Hb S: β 6(A3) Glu → val
- Common to black population
- The effect of alteration from glutamic acid to
2. DELETION
valine is a negative charge wherein glutamic is
- some amino acid is deleted
highly negatively charge compared to valine thus
- reason of thalassemia
leading to loss of negative charges & Hb
Ex.: Hb Gun Hill: (β91 to β 95) → 0 becomes less negative
- In electrophoresis in cellulose acetate, S
3. FUSION migrates slowly than A & F due to substitution of
- Hb Lepore-Baltimore: (G(1-50) B(86-146)) – caused by amino acid 6
cross over during mitosis resulting to fusion - Inheritance:
o Homozygous (SS) – sickle cell anemia
4. ELONGATION/ ADDITION o Heterozygous (AS) – sickle cell trait
5. Hb CONSTANT SPRING (a+31c)
EFFECTS OF HB S:
REASON FOR DELETION & ELONGATION: mutation in  Blacks who are suffering from this are
genetic code protected from being infected from
developing severe falciparum malaria
REASON OF HEMOGLOBINOPATHIES: mutation of gene (Resistance). Hb C, E, B-Thalassemia & G-6PD
NOMENCLATURE deficiency also confer protection in terms of
resistance but not immunity meaning these
1. COMMON NAME
patients may still be infected but may not
a. Morphology- sickling Hb (HbS)
proceed to chronic stage.
b. Place where they were discovered- (Hb
o Falciparum are intracellular species &
Boston) when present in RBC & when RBC is
2. SCIENTIFIC NAME sickle the parasite inside also dies
- Gives structural defect of Hb does no complication arise
- Example: Hb Hb S: β 6(A3) Glu → val o Falciparum metabolizes Hb, when Hb
B: affected polypeptide chain crystalizes it forms into a crystal &
6: sequential amino acid number(s) affected falciparum may no longer metabolize
(primary structure) the Hb
A3: Helix number involved (secondary structure: o RBC w/ falciparum will be
A-H); not necessary as long as primary structure phagocytosis before it causes severity
is indicated
Glu: nature of the abnormality (substitution, HOMOZYGOUS (SS) SICKLE CELL ANEMIA
deletion, addition or globin chain fusion) - Severe anemia because the patient does not
PRIMARY STRUCTURE synthesize the normal Hb A

- sequence or arrangement of amino acid NOTE:


On electrophoresis:
NOTE: o Hb S – 80 -90%
Hemoglobinopathies commonly affects the B chain. o Hb F – 10 -20 %
o Hb A2 – Normal

- Increased RDW
- Retarded growth & sexual maturation & patient NOTE:
suffers sickle cell crises OTHER SICKLING HBS:
- Main cause of Sickle Cell Crises: sickling o Abnormality: Hb S: β 6(A3) Glu → val + unique B-
- Normally when Hb S is fully oxygenated, Hb S is substitution
fully soluble o Hb C-Harlem, C-Ziguinchor, S-Travis
- But when Hb S is w/ reduced O2, Hb S becomes
insoluble wherein it polymerizes (change in HEMOGLOBIN C
molecular arrangement) & forms into tactoid -
Composition: B 6 (A3) Glu → Lys
crystals/fluid crystals (elongated & thin, pointed - A beta structural defect, where glutamic acid is
at both ends) leading to sickling & rigid cell substituted w/ lysine
membrane thus it is no longer adjustable leading
to vassoocclusion thus it cannot easily pass HOMOZYGOUS: Hb C DISEASE (CC)
through the small circulations - N/N in size & shape anemia with numerous
- Sickling is reversible but repeated sickling leads target cells (bull’s eye)
to permanent sickling due to permanent damage - Hb CC crystals formed when deoxygenated &
to RBC membrane dehydrated & the crystals are hexagonal or rod-
- Causes vasso-obstruction shaped crystals or bar of gold shape & blunt ends
- Vasoocclusive crises occurs commonly on small & does not deform the cell meaning it retains its
circulations like in the fingers & toes thus hand- shape but becomes rigid
foot syndrome / dactylitis (inflamed or swollen,
HETEROZYGOUS: Hb C TRAIT (AC)
cyanotic due to absence of circulation) occurs
- RBCs – slightly hypochromic
- Vassoocclusion also occurs in bone & joint
- Less abnormalities
causing pain
- Splenic circulation is also minute, since spleen HEMOGLOBIN SC DISEASE
also serves as a filter of blood thus its circulation - Signs & symptoms are milder than SS but more
is too small, that only deformable red cell may severe than sickle cell
pass through. Due to increase activity of spleen - Electrophoresis: C = S = 50 – 50%
& vassoocclusion, spleen also enlarges - Cytoplasm appears folded like a pocketbook cells
(splenomegaly) causing sequestration of more (RBC w/ cytoplasm folded)
blood leading to hypovolemic shock. When - Hb SC easily crystalize forming a fingerlike
spleen is enlarged & bone marrow is affected projection w/ more than 1 protrusions where
thus both are abnormal in function leading to one is long & protrude away. It also in the form
decrease blood production to the marrow of hand in gloves crystals or pointing – finger or
(aplastic crises) thus the immune system is Washington monument crystal
defective & the patient becomes prone to
infection (infectious crises) as a result of all of OTHER ABNORMAL HEMOGLOBINS
this organ damage occur (marrow lungs) a. Hb METHEMOGLOBIN
- The no. 1 cause of death is infection & in children - M-Saskatoon, Boston, Iwate, HydePark,
the most common is S. pneumoniae. Milwaukee = all have different amino acid
substitution but the effect is the same & not
NOTE: protected from oxidation
SICKLE CELL TRAIT (AS) - Methemoglobinemia w/ congenital cyanosis due
• Predominant Hb is A, only 30-45% is Hb S to overwhelming oxidation even at baby stage &
• Patient usually have no symptoms & sickling is the blood appears chocolate brown
uncommon to occur unless in cases of extreme tissue - Easily precipitate & forms into Heinz bodies
hypoxia because S is minimal in concentration
-
NOTE:
b. HbS W/ ABNORMAL AFFINITY TO OXYGEN
Hb S – Thalassemia:
1. Hb w/ Increase Affinity
o Hb S-A
o Hb S-B – more severe - Example: Hb Chesapeake (a 93 Arg → Leu) – affects
affinity of Hb to O2 & O2 will shift to left
o >1/3: RBC is hypochromic
2. Hb with Decrease Affinity  Size: similar to small lymphocyte
- Example: Hb Kansas (B 120 Asp → Thr) – shift to the o Rule: Slight variation in size & shape is
right acceptable but increase in variation will
be abnormal
NOTE:
 Cytoplasm: uniformly pink without inclusions
Abnormal Hb are connected by weak bonds thus they
 Average lifespan: 120 days
easily precipitate & flocculate
o <120 days: RBC undergoes pre-mature
hemolysis
TEST FOR UNSTABLE HB  Chromium 51 (Cr51): used to measure the
1. ISOPROPANOL PRECIPITATION TEST effective survival of RBCs in vitro
- When heated, 17% Isopropanol at 37 C normal o Half survival time range: 25-32 days
Hb do not precipitate but few will do & unstable o <25 days: RBC undergoes hemolysis
Hb will easily precipitate. o Process: Collect blood from the patient.
- Purpose of Isopropanol: weaken the bonds Radioactive Cr is added on the blood.
Chromated RBC are infuse back
2. HEAT DENATURATION/INSTABILITY TEST intravenously. The disappearance of
- At 50 C for 3 hours, normal Hb will not flocculate chromated RBC will be measured by
but unstable Hb will performing blood cell count in a gamma
ray for every 1 – 2 days for 10 – 14 days.
3. HEINZ BODY STAINING Cr is eluted from circulation 1% a day, its
- If ppt & flocculation happens in vivo forming disappearance is shorter
Heinz bodies which are not easily seen in normal o Purpose: label RBC in order to be
wrights blood smear differentiated from unlabeled RBC
- Supravital staining is employed (brilliant cresyl
blue & crystal violet)
NOTE:
RED BLOOD CELLS  Half – life: no. of days wherein at least 50% of
CHARACTERISTIC OF A NORMAL RBC: radioactivity of Cr or gamma ray disappear
from circulation
 Shape: Biconcave disk  Eliptocytes are usually utilize for thinner &
o Purpose: deformed cells due to acquired condition
 Promotes gas exchange while ovalocytes are egg shape & not thin,
between CO2 & O2 which are often seen in hereditary type.
 For RBC to be flexible &
deformable in order to squeeze
through the microcirculations or RBC MEMBRANE
else it will be prone to lysis. - underlie the lipid layer & regulate shape &
 Diameter: 7 – 8 um deformability because if the membrane is rigid,
o <7: microcyte the cells cannot reshape itself & not flexible
- surrounds the cellular cytoplasm or internal
o >8: macrocyte
environment
 Thickness (MCAT): 1.5 – 2.5 um
- selective transport membrane: separates the
 Average volume (MCV): 90 fL
internal environment to external
 Average Surface Area: 160 um2
o does not easily allow diffusion of
 Shape & Deformity: Flexible & deformable
substances
o Responsibility of: shape, integrity of cell
o It selectively allows the entry of water &
membrane & biconcavity.
electrolytes, inlet & exit
 Mature: anucleated w/ a dense outer rim
- consist of a phospholipid bilayer
 Central pallor: 1/3 of its diameter, it is pale due
to lack of Hb
RBC MEMBRANE COMPOSITION: liquid environment with hydrocarbon tail which
1. 50% PROTEINS is hydrophobic meaning it does not interact with
watery environment. The polar heads are
a. INTEGRAL PROTEINS exposed to the watery environment while the
- crosses phospholipid bilayer hydrocarbon tail is facing inwards because if
- high amount of sialic acid for imparting negative outwards it does not interact with watery
charge to maintain zeta potential environment.

NOTE: KINDS:
Zeta potential 1. EXTERNAL SURFACE: phosphatidyl choline,
- RBC repel each other due to negative cloud glycolipids & sphingomyelin
surrounding them caused by sialic acid. When 2. INTERNAL SURFACE: Cephalin,
zeta potential becomes altered this result to phosphatidylinositol & phosphatidylserine
rouleau formation.
- Component: Glycophorin A & component a
(band 3) b. CHOLESTEROL
- Function: serves as membrane protein - Amount depends upon the concentration of
mechanism in order to allow passage of plasma cholesterol, bile acids & activity of the
molecules either inward or outward enzyme LCAT
- Maintains regulation of membrane fluidity &
permeability to electrolytes. It also maintains the
b. PERIPHERAL PROTEINS
surface area volume of RBC
- Component (cytoskeleton): Spectrin & Actin - Water content of RBC depends mainly on its
- found facing the inner or cytoplasmic ionic gradient wherein the greater the ionic
environment or facing outwards found on the gradient, RBC becomes hypertonic thus water
outer or inner surface enters RBC & too much entry of water causes
- adds up to the adherence of lipids swelling of RBC & eventually burst & decrease
MEMBRANE ENZYME SYSTEM FOUND OF THE SURFACE surface area volume
OF RBC:
AMOUNT DEPENDS UPON THE CONCENTRATION OF:
- Form the Active Transport System 1. PLASMA CHOLESTEROL: if plasma cholesterol
is high, patient has hypercholesterolemia
a. Na/K ATPase (liver diseases) macrocytes & target cells are
- Regulates entry of Na & K in & out of the cell in present, because of loading of some
cholesterol into the cell membrane causing
order to maintain Na as a major extracellular
the cell to widen
cation while K is the major intracellular cation
2. BILE ACIDS – for solubilization
thus when it is impaired there would be 3. Activity of the enzyme LCAT
excessive inlet & outlet of cations. If excessive
cation entered, this will result to hypotonicity of
RBC causing now the entry of water, swelling & 3. 10% CARBOHYDRATES
lysis of RBC - Some serve as red cell antigens (e.g. ABH antigen
on surface of RBC)
b. Ca/Mg ATPase - Often in combination w/ lipids (glycolipid) or
protein (glycoprotein)
2. 40% LIPIDS - Ex.: Glycophorin A, B , C/D
a. PHOSPOLIPIDS ABH – blood antigen ABO – blood
- Serves as a liquid sealer, because it is liquid group Ab
stabilized by proteins where they are connected
NOTE:
- Arrangement serves as liquid sealer wherein
 A-spectrin, Ankyrin, Band 3, B-Spectrin,
there is 2 phospholipid layers containing a
Protein 4.2 & 4.1 & Glycoprotein are
phosphate head which is polar thus it reacts with
important in maintaining the shape & size of - Main function: generates NADPH and reduced
RBC & abnormality of these will lead to glutathione (GSH) in the presence of Glucose 6-
deformation of cells. PO4 dehydrogenase (G6PD). GSH protects the
 Defective A-spectrin, cell will be spherocytes hemoglobin from oxidation by peroxides.
or pyropoikilocytes - Process: If G6P converts into 6-
phosphogluconate, it produces NADPH & the
METABOLIC ACTIVITIES IN THE RBCS enzyme used is G6PD & the substrate is NADP
- RBC is metabolically active when it is maturing which is reduced into NADPH which may
because when it becomes mature it does not become H donor wherein it transfers the H to
have nucleus & mitochodria ferric iron & ferric becomes ferrous. It also
maintains the glutathione in the reduced form
RULE: which also serves as a reducing agent wherein it
The more immature the RBC, the more metabolically detoxifies the toxic radicals (H2O2 – strong
active it is which is related to the amount of the RNA oxidizing agent causing oxidation of lipids & Hb
which indicates the cellular immaturity & metabolic & this is detoxify into H2O & O2). Glutathione
activity. also brings back the ferric form of iron into
ferrous.
1. EMBDEN MEYERHOF PATHWAY/GLYCOLYTIC
PATHWAY 3. RAPOPORT-LUEBERING SHUNT
- Since RBC are already devoid of mitochondria, - Generates 2,3-DPG /2,3-BPG which regulates
glycolytic pathway goes anaerobic. the affinity of Hb to O2 by lowering the affinity
- Main Function of Anaerobic Glycolysis: promoting release of O2 causing a shift to the
production of the cells energy which is ATP right
- Process: for every glucose metabolize, it will
produce 2 ATPs for the glyceraldehyde-3- 4. METHEMOGLOBIN REDUCTASE PATHWAY
phosphate & for 2 ATPs for Dihydroxyacetone - Reduction of methemoglobin by NADPH is more
Phosphate thus a total of 4 but the net energy efficient in the presence of methemoglobin
utilized is a net of 2 reductase (cytochrome b5 reductase) which
- Major source of red cell’s ATP (90-95%) serves as an intermediate electron carrier. It also
prevents oxidation of heme by reducing ferric to
ferrous with the use of NAD.

CONNECTIONS OF THE 4 PATHWAYS


- EMBDEN MEYERHOF PATHWAY is an anaerobic
& goes as anaerobic glycolytic pathway, mainly
for the production of 90-95% of RBC ATPs. The
HEXOSE MONOPHOSPHATE SHUNT is important
for the production of NADPH & GSH for the
maintenance of Hb iron in the reduced form
because it can reverse the ferric into ferrous.
GSH can also detoxify H2O2 converting it into
H2O & O2. RAPOPORT-LUEBERING SHUNT is
necessary for production of 2,3 DPG which
2. HEXOSE MONOPHOSPHATE SHUNT/PENTOSE maintains affinity of Hb to O2.
PHOSPHATE SHUNT METHEMOGLOBIN REDUCTASE PATHWAY
- Arises from Embden Meyerhof Pathway but it produces NADH which serves as reducing
undergoes oxidative glycolysis providing 5 – property & may also donate its hydrogen to
10% ATP cause reduction of ferric into ferrous.
- Contains the major anti-oxidant of cells
BREAKDOWN OF RBCs - Process: Hemoglobin is released into the plasma
- As RBC ages, there is a decrease in its enzymes and can be filtered by the kidneys. Plasma
causing slow reaction, thus causing a decrease in haptoglobin and hemopexin salvage the
ATP production & metabolic activity decreases & released hemoglobin so that its iron is not lost in
everything also decreases causing now the urine
distortion thus if more ions enter the RBC, more - Ex.: when RBC is not deformable they undergo
water will enter causing now swelling and size intravascular hemolysis (spherocytes)
also decrease and an increase in density.
- This natural deterioration leads to its
MECHANISM TO CONSERVE THE HB:
phagocytosis. When cells are deformed it will be
1. HAPTOGLOBIN
recognized by splenic macrophages as a damage
- synthesize by the liver & released into the
or senescent RBC wherein spleen filters blood to
plasma
removed old & damage cells (splenicullin), this
- Function: combine w/ hemoglobin preventing
process happens extravascularly. Approximately
Hb from being filtered by the kidneys &
1% of RBC leave the circulation each day & are
hemoglobin-haptoglobin will be brought into the
broken down by mononuclear phagocytic
liver for metabolism. Hb will undergo breakdown
system
by separating heme & globin. Globin will be
recycled as amino acids, & goes back to amino
a. 90% EXTRAVASCULAR (MACROPHAGE-
acid pool. Heme is recycled as iron is
MEDIATED)
immediately utilized or stored as ferritin but
- destruction of senescent red cells by splenic
haptoglobin cannot repeat the function, it will
macrophages
carry Hb to the liver but it cannot repeat its
- release of Hb occurs inside the macrophages &
function, it cannot go back. Thus, in increase
unconjugated Hb is released in the circulation
hemolysis haptoglobin is also depleted.
- Process: Hemoglobin undergoes degradation
- In short: haptoglobin binds w/ free Hb thus
within the macrophage where iron is stored as
forming hemoglobin-haptoglobin & delivered to
ferritin, amino acids of globin are returned to
the liver
the metabolic amino acid pool and
protoporphyrin is converted to bilirubin which
2. ALBUMIN
is released into the plasma and excreted by the
- Process: Hb in plasma becomes oxidized into
liver in bile.
methemoglobin which combines with albumin
forming methemealbumin, this will be brought
b. 10% INTRAVASCULAR DESTRUCTION
into the liver for processing.
(MECHANICAL HEMOLYSIS)
- In short: albumin bind to methemoglobin
- may be caused by the turbulent environment in
forming methemealbumin, & increases in
the circulation
hemolysis due to increase formation
- lysis occurs in the circulation thus release of Hb
is directly in the plasma & increase in
intravascular hemolysis can cause the RBC to
appear pink. Hb is a low molecular weight
protein that can be easily filtered by the kidneys
& be excreted into the urine causing
hemoglobinuria making urine red or pink &
when Hb stayed in room temp. for prolonged
time it will be oxidized into methemoglobin.
When Hb is excreted, it will lose the components
(60% Fe, Protoporphyrin 9, globin & amino acids)
but the body has a compensatory mechanism to
conserve the Hb.
LABORATORY EVALUATION OF RBC
 RBC count
 Hemoglobin
 Hematocrit
 RBC Indices
 Peripheral Blood Examination
o Blood Smear for differential counting &
morphology examination of RBC (PBS)
 Reticulocyte measurement
 Osmotic Fragility test
 Erythrocyte Sedimentation Rate

CRITERIA FOR PATHOLOGIC RBC


 Size (7-8 um)
 Variation in size (80-100 fL)
 Area of Central Pallor (1/3 & clear)
3. HEMOPEXIN
 Cytoplasm (clear w/out inclusion)
- Process: Hemopexin combines w/ heme. When
heme is separated from methemoglobin it can
be still conserve by binding of hemopexin to
heme forming the hemopexin complex & will be
brought to the liver
- In short: heme bind with hemopexin forming
heme-hemopexin complex

NOTE:
Whether intravascular or extravascular, iron is
recycled.

ANISOCYTOSIS
 increase in variation in size thus increasing RDW a measure of variation in size
 Refers to variation in color after staining which is related to the hemoglobin content of the cell, since it is the
hemoglobin that takes up the stain wherein the appearance of the cell after staining is directly proportional to
the hemoglobin content
CELL TYPE MORPHOLOGIC APPEARANCE DEFECT OR CHANGE ASSOCIATED CONDITIONS
NORMOCYTE Biconcave disc  Normal
 Not all cells are normal there is a classification
known as normocytic anemia which are normal
in size (MCV: 80-100) but there is anemia which
is related to the decrease in production
MICROCYTE  Smaller RBCs  Main cause: Any defect in heme or globin that
 Diameter: <7 µm results in IMPAIRED HB SYNTHESIS thus cells
 MCV= <80 fL undergo extra division & the cells hardly
reached its optimum MCHC
Note: It is efficient to use MCV  Develops from:
because diameter depends on the o ineffective iron utilization, absorption,
smear prepared. or release
o decreased or defective globin synthesis
 Diseases caused:
o IRON DEFICIENCY ANEMIA: affects
heme
o SIDEROBLASTIC ANEMIA (heme) – iron
precipitate due to protoporphyrin 9
deficiency leading to siderotic granules
& its occurrence in RBC appears as
sideroblast
o THALASSEMIA (globin)
o ANEMIA OF CHRONIC
INFLAMMATION/DISEASE:
inflammation activates macrophages &
T lymphocytes causing increased
production of cytokines that will inhibit
normal iron metabolism thus affecting
heme synthesis
o LEAD POISONING – lead inhibits
enzymes (heme synthase) in heme
synthesis
MACROCYTE  Larger than 10 µm CAUSES:
diameter due to failure of a. ACCELERATED ERYTHROPOIESIS – fast RBC
mitosis prod. Until faster release of reticulocyte in
 Diameter: >9 µm circulation as a compensation to blood loss or
 MCV= >100 fL lysis. If patient suffers from blood loss or lysis,
blood cell count decreases causing hypoxia &
the kidneys will produce increase
erythropoietin causing acceleration of
erythropoiesis thus the bone marrow will
release even immature reticulocytes
(stress/shift reticulocytes) thus it is larger in
size
b. DEFECTIVE DNA SYNTHESIS – affects nuclear
maturation resulting to nuclear arrest which is
prolonged mitosis due to immaturity of nucleus
which is the central control of the cell due to
deficiency in DNA synthesis thus it undergoes
slow maturation & mitosis leading to large
cells; failure of division
c. IN CONDITIONS WHERE MEMBRANE
CHOLESTEROL AND LECITHIN LEVELS ARE
INCREASED – cell membrane is loaded w/
cholesterol thus increasing cell membrane &
the cell due to hypercholesterolemia this is due
to disturbance in the lipid conc. of plasma &
may be seen in obstructive liver disease
wherein liver is the main organ that
metabolizes fats and where VLDL came from.
Obstructive liver disease leads to incomplete
metabolism of fats thus patients suffer from
hypercholesterolemia.

 HEMOLYTIC ANEMIA & ACUTE BLOOD LOSS –


bone marrow compensates for the blood loss
or lysis by producing immature RBC thus high
reticulocyte count
 MEGALOBLASTIC PROCESS – affected by
impaired DNA synthesis because cells fail to
divide or suffer from long mitosis resulting to
oval macrocytes
 CHEMOTHERAPY – the chemical infused to
destroy the malignant cells often target the
DNA of cell thus affecting DNA
 LIVER DISEASE - increase cholesterol lipids
causing loading to phospholipid bilayer
 HYPOTHYROIDISM – thyroid is necessary for
metabolism, T3 & T4, for regulating other
hormone
 POST-SPLENECTOMY – not only macrocytes
persist in circulation but also all cell
abnormalities. The spleen filters 5 – 10% of
blood allowing inclusions & abnormal cells to
be removed thus removing the spleen will lead
to no filtration of blood causing abnormality in
circulation.
 ALCOHOLISM
DINMORPHIC RBCs  Presence of 2 distinct populations of red cells that may differ in size, shape or
hemoglobin content thus it may be a mixture of hypochromic cell & normochromic
cell or a microcytic cell or macro
 SEEN IN:
o ANEMIA AFTER TRANSFUSION – after transfusion, on the examination of
patient’s blood smear, there is 2 population of cell, the patient RBC &
transfused RBC, the cells are pale (hypochromic) & microcytic, mixed w/ the
transfused blood (normocytic & normochromic)
o IRON DEFICIENCY DURING THERAPY – cells are microcytic & hypochromic
o IRON DEFICIENCY & VIT. B12 – Iron stored in bone marrow, but storage of
iron is not proportional where the other parts are proportional while the
others are not, thus the developing cells near w/ iron develops as normocytes
while cells w/ developing iron develops as microcytes thus it is a mixture of
microcytic & normochromic cells
o COMBINED VIT. B12 /FOLATE – macrocytosis, 2 most common causes of
megaloblastic anemia DEFICIENCY AN IN SIDEROBLASTIC ANEMIA –
microcytosis, normocytic cell ; a group of anemias that show different
morphology

ANISOCYTOSIS & POIKILOCYTOSIS MACROCYTOSIS GRADING (no


GRADING variation)
Percentage of RBC differ in size or shape from normal RBCs.
Normal 5% 1+ (slight) 25%
Slight 5-10%
1+ 10-25% 2+ to 3+ (moderate) 25-50%
2+ 25=50%
3+ 50-75% 4+ (marked) >50%
4+ >75%
Used if cells vary in size and shape
ANISOCHROMASIA
 variation in the hemoglobin concentration
 absence of uniformity of color
 directly relates to Hb
 Color of cells when stained depends on the Hb
CELL TYPE MORPHOLOGIC APPEARANCE DEFECT OR CHANGEASSOCIATED CONDITIONS
NORMOCHROMIC Homogenous with central Normal
pallor (1/3)
Measure: MCHC
HYPOCHROMIC  RBCs show central DEFECT IN HB SYNTHESIS (HEME/GLOBIN) DUE TO:
pallor (exceeds 1/3 of  decrease in heme synthesis
the diameter of the red  defect in globin chain synthesis
cell)  Iron Deficiency Anemia (IDA)
 Thalassemia
ANULOCYTE/ GHOST  Thin & poorly
CELLS hemoglobinized cells RULE: Not all hypochromic cells are microcytic but all
- very hypochromic (anulocyte) microcytic cell are hypochromic
cells
ANISOCHROMASIA & POIKILOCYTES
SPHEROCYTIC Cells that lack central pallor  Defects in the cytoskeletal proteins
and with reduced diameter  Hereditary and acquired spherocytosis
TARGET CELLS  Central pale area is
filled with hemoglobin
resulting to bulls eye &
further surrounded by
thin rim of Hb
 On side view, there is
protrusion on the
center
POLYCHROMASIA PROBLEM: difference in RNA & Hb among RNA & Hb
 Refers to the display of different shades of color which is Diffused bluish-gray tint of
RBCs & is not directly related to Hb content
 Already fully hemoglobinized but the display of different colors is caused by the
differences in the affinity of the cellular component (Hb & rRNA) wherein Hb takes
up the acidic dye & the rRNA takes up the basic dye
 Exemplified by the fifth stage of erythropoiesis: reticulocytes which is the first non-
nucleated stage but it has remnants of RNA & is fully hemoglobinized

RULE: The more immature, the more RNA it has thus the more chromatophilic it is.
 Not directly related to Hb content but on the RNA content
 The reddish or orange color caused by eosin of Hb & the bluish color stained by
methylene blue of rRNA will be mixed in the cell & the colors produced is mixed
resulting to diffuse coloration of cells
 Increase polychromatic cells indicates immature cell (reticulocyte) which is
synonymous to reticulocyte

APPEARANCE: diffuse coloration, no central pale area


RETICULOCYTE IDENTIFICATION:
 Supravital stains (new methylene blue & brilliant cresyl blue) – appropriate staining
of living reticulocyte & the cells will appear as larger cells w/ granules (RNA).
 Supravital stains precipitate the RNA thus they appear as dark blue precipitate
against pale blue cytoplasm. The granules appear in a network or reticulum thus
called reticulocytes

HYPOCHROMIA GRADING POLYCHROMASIA GRADING


1+ Area of central pallor is ½ of cell diameter Slight 1%
2+ Area of pallor id 2/3 of cell diameter 1+ 3%
3+ Are of pallor is three-quarters 2+ 5%
4+ Thin rim of hgb 3+ 10%
4+ >11%

1+ (slight) Central pallor: 1/3 ro 2/3 of cell’s diameter 1+ 1-3 polychromatophilic cells/ field
2+ to 3+ More than 2/3 2+ 3-5 PcC/f
(moderate)
4+ (marked) Thin rim on the periphery 3+ >5 PcC/f

VARIATION IN SHAPE OF RBC


POIKILOCYTOSIS- increase variation in shape ; named after their shape
PROTEINS – maintain shape & size of the cell, preventing it from collapsing
LIPIDS – main fluid barrier

 Abnormality in proteins, lipids & carbohydrates affects the shape of the cell
CELL TYPE MORPHOLOGIC DEFECT OR CHANGE DEFECT OR CHANGE ASSOCIATED
APPEARANCE ASSOCIATED CONDITIONS
CONDITIONS
OVALOCYTES  Range from egg- HEREDITARY OVALOCYTES:
(ELLIPTOCYTES) shape, slightly oval ELLIPTOCYTOSIS: (25-  Myelodysplastic syndrome
to sausage, rod, or 90%)  Thalassemic syndrome
pencil forms  decrease in  Megaloblastic syndrome –
 Despite of their skeletal megaloblastic
shape they function membrane ELLIPTOCTE:
normally depending CHON band 4.1  Hereditary elliptocytosis
on their hemoglobin  increase heat  Idiopathic myelofibrosis
content, flexibility & sensitivity of  Small numbers may be
deformability spectrin acquired in:
 Some appear o iron deficiency,
normochromic or ACQUIRED: 10% o megaloblastic
hypochromic  No problem or anemia,
meaning they are deficiency o myelophthisic
abnormal in shape affecting the anemia,
but normal Hb protein o thalassemias &
content thus membrane but sickle cell anemia
function normally the cell
 Some are became
normocytic but distorted
elongated sized but because of
the MCV is the different
same. Some are oval condition
& macrocytic
(macroovalocyte)
which is a significant
observation related
to megaloblastic
anemia
 Hb appears to be
concentrated at the
two ends of the cell,
leaving a normal
central pallor area.

NOTE: Membrane
abnormality for both is the
same
NOTES:
Peripheral proteins can be found on the surface or inner surface underlying the phospholipid layer, these proteins are
connected to the transmembrane proteins & phospholipid bilayer which adds on the integrity thus deficiency in
spectrins, protein 4.1, 4.2 will alter the membrane thus deformed.
SPHEROCYTE  sphere & not  indicates a TWO MOST COMMON CAUSES:
biconcave hemolytic  HEREDITARY
 Smaller in diameter process SPHEROCYTOSIS/INTRINSI
than normal RBC & (hemolysis C MEMBRANE DEFECT –
w/ smaller volume results from a protein or spectrin
because it is fully membrane abnormality, deficiency in
distended due to abnormality) ankyrin or protein band 3.
increase hb thus it cannot Spectrin dimer is
 On side view it has a enter onto the connected to the integral
small biconcavity microcirculatio protein, ankyrin underlies
but very small not n & as they the phospholipid bilayer,
enough to make enter they these add to the integrity
them flexible or immediately of phospholipid bilayer
deformable burst or lyse thus a deficiency in these
 With concentrated protein will make the
hb content phospholipid bilayer,
 No visible or weaker, & some of portion
decrease central of phospholipid will be
pallor easily detached into the
form of microvesicles.
IN SHORT:
A deficiency in spectrin, ankyrin &
protein band 3 leads uncoupling of
phospholipid layer & some of lipids
will be easily detached into the
form of microvesicles. Repeated
detachment leads to a smaller cell
having no central pallor,
spherocyte.

OTHER EXPLANATION:
Weak membrane causing weak
dissociation of deficiency in
cytoskeletal protein

 AUTOIMMUNE
HEMOLYTIC ANEMIA: Ab
to the red cell will
attached to the red cell w/
their FC portion exposed
which will be recognized
by macrophages that have
Fc receptors on their
receptors & they pit off.
Macrophages pull off Ab
attached on RBC but as it
pulls off, it removes
portion of RBC. The
remaining fragment of RBC
will reseal to form a
smaller cell w/ a filled
volume. Repeated pitting
may occur resulting to
microspherocyte.

SPLENIC CULLING – removal of old


& damage red cell
PITTING – process of removing
inclusions & Ab attached to RBC

OTHER MECHANISMS (LOSS OF


MEMBRANE):
 NORMAL AGING PROCESS:
lose enzymatic properties
& will lack ATP caused by
decreased in enzymatic
activity of enzymes involve
in Embden Meyerhof
pathway resulting to lack
of ability to maintain their
shape & size. Na/K ATPase
needs ATP for activity thus
when cell lacks ATP, this
can no longer be
maintained & transport of
Na/K is compromised & as
an effect the cell can be
crenate or distorted.
 STORAGE PHENOMENON
– after transfusion of
stored blood, patient
blood smear shows lots of
spherocytes but no
abnormality in the
membrane but the cells
undergo change in their
shape due to storage
lesion. During the storage
of blood in blood bag, it
will be consumed, the cell
will lack glucose to
produce ATP & the main
source is glycolysis, but as
the cell lack glucose this
will lead to lack of ATP
production.
 SEVERE BURNS:
Since phospholipid is a fat,
it is easily dissolve by heat
thus will fragment &
vesicle out of phospholipid
bilayer resulting to
spherocyte & repeated
fragmentation leads to
microspherocyte.
BURR CELL  Microscopically burr  Depletion of BURR CELL
& echinocyte ATP 1. Occur in situations that
appear similar.  Exposure to cause a change in tonicity
ECHINOCYTE hypertonic (hypertonic) of the
 Sea urchin solution intravascular fluid plasma
 Crenation occurs in seen in dehydration &
vitro azotemia related to uremia
 An artefact &
insignificant which 2. ASSOCIATED W/ RENAL
may be seen on INSUFFICIENCY - wherein
thinner part of the kidneys important in
cell removing metabolic waste
 With evenly in the blood, it also filters
distributed, blood but do not remove
uniformly sized 10- abnormally shaped cells &
30 blunt spicules or inclusions. It functions to
bumps remove metabolic
products, dissolve
RULE: Both reactions are chemicals & substances
reversible (salts, acids, & excessive
water). In renal
BURR CELL insufficiency the kidneys
 Very clinically cannot function, resulting
significant which to non-removal of
indicates changed in metabolic products such as
the plasma tonicity the non-protein
 Microscopically the nitrogenous substances,
appearance of the blood urea nitrogen &
spicules is the same creatinine, these
w/ echinocytes thus substances builds up in the
relate in conditions. blood altering the tonicity
 Crenation occurs in thus cells will crenate
vivo while still in the
 Reversible in intravascular fluid.
formation, when
patient underwent RULE: The no. of burr cell indicates
dialysis thus NPN is the severity of renal damage.
cleared out from the
plasma maintaining ECHONOCYTES
the normal tonicity 1. Several hours old
of plasma anticoagulant blood stored
 With irregularly at room temperature
sized & unevenly causing an artefactual
spaced 10-30 blunt effect due to prolonged
spicules storage at prolonged
temperature
2. Stored blood causes
crenation of cell due to
depletion of ATP &
biochemical activities

RULE: Both reactions are reversible


ACANTHOCYTES  Irreversible  May be caused  ASSOCIATED W/ END
formation by changes in STAGE LIVER DISEASE –
 No longer a disc the ratio of liver is main organ which
shaped, with few (3- plasma lipids. metabolizes & stores fats
12) irregularly Since cell such as cholesterol &
spaced, pointed membrane is lipoproteins but when it is
spicules/thornlike made up of 2 diseased it will not be able
projections of layers of to metabolize the different
various lengths and phospholipids phospholipids &
widths + cholesterol cholesterol resulting to
 Indicates that maintain abnormal ratio in plasma.
permanent the When there is
membrane damage, permeability of hypercholesterolemia due
some are long & membrane to to liver disease this may
short thus it the passage of lead to abnormal ratio of
indicates that the ions. plasma lipids leading to
cell can no longer  Changes in distortion of RBC into
regain its normal cholesterol, acanthocyte.
shape lecithin &
 sphingomyelin NOTE: The conc. of cholesterol
depends on the conc. of plasma
cholesterol, activity of LCAT & bile
acids.

 ALCOHOL INTOXICATION –
affects liver leading to
hepatic cirrhosis, the
alcohol has a direct effect
on RBC
 ALCOHOLIC CIRRHOSIS
 PK DEFICIENCY – pyruvate
kinase is involved in EMP,
it catalyzes the conversion
of phosphoenolpyruvate
to pyruvate wherein ATP is
produced thus deficiency
in PK leads to deficiency in
ATP which is important in
maintaining cell
membrane
 CONGENITAL
ABETALIPOPROTEINEMIA
– absence of
betalipoprotein which is
related to the inability of
the liver to metabolize it.
LDL is necessary but must
not be in excess thus the
absence of betalipoprotein
leads to red cell distortion
 VITAMIN E DEFICIENCY –
Vit. E is needed for
preventing peroxidase
oxidation of the cell
membrane
POST-SPLENECTOMY – removal of
spleen, all abnormalities will be
seen in the circulation
STOMATOCYTES  Normal sized cell Membrane defect that COMMON CAUSES: HEREDITARY
(MOUTH CELLS) with an elongated results in high cellular  HEREDITARY
HYDROCYTE or a slit like mouth sodium and low STOMATOCYTOSIS – main
or stoma of central potassium content problem in the transport
pallor of Na/K. Due to defect in
 cup-shaped on SEM the transport of Na/K,
 fully hydrated cell there is high cellular Na
due to defect in the thus water enters into the
transport of Na/K cell & the cell becomes
ATPase hydrated & low K
 swollen cell due to  RH NULL DISEASE – second
increase water major blood group (rhesus
content blood group), the Ag in this
blood group are the capital
D indication of positive or
negative & also C & E. Rh
null means no D, C & E Rh
Ag located on the cell
membrane. Rh Ag are
integral part of cell
membrane for the
transport of molecules
thus absence affects the
maintenance of RBC thus
transport of Na/K is
altered leading to
stomatocytosis

ACQUIRED CASES: MOST


COMMON CAUSE:
 Obstructive liver disease
 Alcohol excess or
intoxication
LESS COMMON:
 myelodysplastic
syndromes
 hydroxyurea therapy –
therapy for leukemia
 Hereditary stomatocytosis
TARGET CELLS  Has a bulls eye, w/ a  Increase in HB DEFICIENCY:
(CODOCYTES)/ button or cholesterol &  Hemoglobinopathies
PLATYCTES/LEPTOCYT concentrate Hb thus phospholipids  Thalassemia
E a dark center  Excess surface  Hemoglobin C Disease or
followed by clear Hb area to volume Trait
area & followed by ratio  Iron deficiency anemia
thin rim of Hb  Any hemoglobin
 May have or have abnormality
not clinical
significance (Hb OTHERS:
deficiency)  Liver diseases – affects
 Large cell w/ small level of plasma cholesterol
Hb as compared to  LCAT deficiency –
its volume or size hypercholesterolemia,
causing the some of cholesterol will
redistribution of Hb load causing the cell to be
into a button of Hb larger in size & Hb content
at the center (Iron  Post-splenectomy
deficiency anemia,
thalassemia,
hemoglobinopathies
)
 With a central area
or hb surrounded by
a colorless ring and
a peripheral ring of
hb
 bell-shaped (codon)
or tall hat shaped on
SEM
 resembles a tall
Mexican hat cell on
SEM
 non-specific finding,
seen on several
findings & are
artIfactual in nature:

1. Drying Artefact –
slow drying due to
humid environment
causing Hb to
crystallize at the
center & dissolve
forming a
concentrated Hb at
the center resulting
to a target cell
2. Decrease Hb

PLATICYTES - On front view,


it appears as a flat cell
LEPTOCYTE – remains
connected w/ the peripheral
Hb but clinical significance is
the same
NOTES:
ACANTHOCYTE: abetalipoproteinemia TEARDROP CELL: myelofibrosis
BURR CELL: renal disorder TARGET CELL: non-specific

POIKILOCYTES SECONDARY TO TRAUMA


EXCESSIVE PHYSICAL TRAUMA IN THE CVS DUE TO:
1. Presence of fibrin strands causing physical trauma to the red cells passing through leading to fragmentation
2. Blood vessels that are abnormally too small for RBC to circulate

TWO PATHWAYS OF FRAGMENTATION ARE RECOGNIZED


a. Alteration of normal fluid circulation (DIC, TTP, MAHA)
b. Intrinsic defects of the red cell (spherocytes, antibody altered red cells, inclusions
CELL TYPE MORPHOLOGIC DEFECT/S OR CHANGE/S ASSOCIATED
APPEARANCE ASSOCIATED CONDITIONS CONDITIONS
SCHISTOCYTES  Very significant FRAGMENTATION DUE TO: Hall mark of Hemolytic
(SCHIZOCYTES) finding indicating a 1. Exposure of cells to anemias
hemolytic anemia abnormal conditions like
heat (affects RBC in CAUSES:
General term for fragments circulation) or mechanical PROSTHETIC HEART
of RBCs varying in shapes: trauma VALVE – damaged on
 Helmet cells 2. Contact with fibrin artificial heart valve will
 Triangular cells strands or damaged exposed artificial
 Keratocytes/Horn endothelium – fibrin components such as
cell deposits & fibrin strands rubber, causing damage
present slicing of RBC to RBC & RBC suffer from
during their passage. If physical trauma leading
the current of blood flow to hemolysis
is strong this will push the
cell to the fibrin strand MICROANGIOPATHIC
causing now HEMOLYTIC ANEMIA &
fragmentation seen in DISSEMINATED
microangiopatic anemia & INTRAVASCULAR
disseminated COAGULATION
intravascular coagulation SIMILARITIES:
3. Intrinsic defects of RBCs – coagulation mechanism
RBC has the problem is activated causing now
 Spherocytes (no more the formation of blood
adjustment in shape clots due to fibrin
& volume), Ab altered threads deposited on the
red cells (recognized circulation causing now
by macrophages & obstruction & limit the
hemolyze them), Red normal circulation
cells containing leading to physical
inclusions (pitting of trauma. If clots are
cell causing formation bigger the more
of bite cell), etc. obstruction which will be
difficult for RBC to pass
thru.

DIFFERENCE:
MAHA – abnormal
formation & deposition
of blood clots occurs in
blood vessels
DIC – affects all vessels
since it is disseminated

CLOSTRIDIAL
INFECTIONS – toxin
produced damages RBC
& activate coagulation to
cause abnormal clot
formation

THROMBOTIC
THROMBOCYTOPENIC
PURPURA & HEMOLYTIC
UREMIA SYNDROME
SIMILARITIES: platelet
aggregates causes
obstruction of circulation
due to abnormal
activation of platelets
wherein when platelets
are aggregated they form
together to form a
thrombus
KERATOCYTES (HORN KERATOCYTE – pair of BITE CELL: Results from Hemolytic anemias
CELL), BITE CELLS, spicule surrounded by a removal of a Heinz body from G6PD deficiency
DEGMACYTES gap a cell by splenic macrophages
BITE CELL – bite (splenic pitting) or when an
appearance erythrocyte is caught on a
 Erythrocytes with a fibrin strand or during pitting.
pair of spicules or  When a cell has an
horns surrounding inclusion it will pit off
a gap in the cell or pull off by
outline macrophages thus
removing a part of
RBC & the cells will
assume a bite cell

1. Heinz inclusion on a cell


will pass through a limited
basement membrane
2. Rigid portion will be left &
if able to pass through it
will become a teardrop
cell but if removed RBC
will reseal & adjust the
volume & shape
SEMILUNAR BODIES  Half-moon / crescent  Frequently observed Overt Hemolysis
cells in malaria which is a
 Large pale pink hemolytic disorder &
staining ghost of the in other conditions
RBC causing overt
 Complete round w/ a hemolysis.
visible crescent shape  As RBC is hemolyze it
of Hb or complete will release its Hb
crescent content & only a small
portion will be left
that is remain
attached on the
membrane indicating
an overt hemolysis
TEARDROP CELLS  Pear-shaped or  Caused by excessive MOST COMMON:
(DACROCYTES) teardrop-shaped squeezing & results to
w/ a single fragmentation during MYELOFIBROSIS W/
elongated point splenic passage MYELOID METAPLASIA –
 One end is  Vessels constricted bone marrow is
rounded the other causing RBC to infiltrated by fibrous
end is pointed squeeze itself which materials, thus there is
resulting to a pear, causes too much overgrowth of fibrous
drop or rocket squeezing resulting to cells thus if bone marrow
shape permanent damage & is unable to produce
 Hall mark of may no longer revert blood cells resulting to
myelofibrosis to its shape thus metaplasia & spleen is
loosing capacity to involve in the production
regain normal shape of RBC which may suffer
also of fibrosis causing a
limitation of its
circulation. Blood vessels
in the spleen are
normally small or minute
but in myelofibrosis the
more they become
limited thus increase
microcirculation causing
teardrop cell which is the
hall mark of
myelofibrosis.

HYPERSPLENISM –
spleen enlarge causing
circulation to be limited

PRESENCE OF INCLUSION
BODIES – makes RBC
rigid like Heinz bodies
which commonly
attached on the
membrane of RBC
making the membrane
rigid & when it passes
through the splenic
circulation which is too
limited, it has to squeeze
itself leading to its:
1. Detached & cell
becomes a bite cell
2. Pulled & be pointed
because it is the last to
exceed the membrane
causing a tear drop shape

MYELOPHTHISIC
ANEMIA – bone marrow
is infiltrated w/ abnormal
cell like in leukemia thus
circulation becomes
limited

UNCOMMON:
Pernicious anemia
B thalassemia
MICROSPHEROCYTES &  Are red cell Results from repeated MICROSPHEROCYTE:
PYROPOIKILOCYTE fragments fragmentation of RBC & from Severe burns – thermal
 MCV 60 fL thermal damage to cell injury on RBC membrane
 Smaller than membrane wherein fats are easily
spherocytes damage causing the
 Also lack central membrane to vesicle, if
detachment is mild it will
result to spherocyte but
if there is continuous
fragmentation, it will
become the largest
fragment, the
microspherocyte

PYROPOIKILOCYTE:
Hereditary
pyropoikilocytosis –
there is a membrane
damage of red cell due to
sensitivity to heat. A
normal RBC fragments at
49 C but cells with HPP
will fragment at low T
(45-46)
OTHER APPEARANCES RELATED W/ HEMOLYSIS: KNIZOCYTES – appear like PYKNOCYTES – distorted
pinched bottle having or irregularly contracted
BLISTER CELL – with thinned portion due to cell caught constriction on one of its part red cells, seen in
on fibrin strands wherein it might have encountered indicating that it might blister hemolytic anemia
along the circulation such the portion of the membrane or bud off wherein the bud
is pull apart. may detached & become
defragment. The remaining
fragment, the larger one may
become a spherocyte or the
smaller being
microspherocyte, seen in
hemolytic anemia normally in
infants but w/ lesser number

POIKILOCYTES & CRYSTALS SECONDARY TO ABNORMAL HEMOGLOBIN CONTENT


CELL TYPE MORPHOLOGIC DEFECT/ CHANGES ASSOCIATED CONDITIONS
APPEARANCE
DREPANOCYTES/SICKLE  Most insoluble of  Polymerization of Sickle cell anemia
CELLS the Hb variants hemoglobin Indicates Sickling Hb
 Fully soluble Hb  Sickling is
when exposed to reversible when
normal O2 but exposed to normal
when O2 but repeated
deoxygenated it sickling causes
becomes insoluble permanent
& crystallizes at damage on the
low O2 tension. membrane &
The crystals are assumes
thin & elongated
with pointed ends
altering the shape
of the cell having a
curved, straight L,
V, & S shape

HB CRYSTAL HB CC CRYSTAL
 With one or more  Hexagonal
fingerlike, blunt-pointed  Both ends are blunt &
projections that protrude rectangular
from the cell membrane  Parallel sides
leaving a pale area at the  Formed within the membrane (RBC) but does not
opposite end deform RBC causing a normal shape but it makes
 Has more than one protrusion & the longest one RBC rigid thus the RBC is not deformable &
protrude away flexible thus prone to hemolysis
 Washington monument, Pointing finger, Hand  Bar of gold
glove  Crystalizes when Hb is O2 or dehydrated
INCLUSIONS: ABNORMAL HEMOGLOBIN PRECIPITATION
REASON:
1. Precipitation of abnormal component in the cell
2. Disturbance or abnormality in the developmental or maturation of RBC (developmental organelles)
INCLUSION MORPHOLOGIC DEFECT/S OR CHANGE/S ASSOCIATED CONDITIONS
APPEARANCE
HEINZ BODIES  Large (1 to 3 Results from  G-6-PDH DEFICIENCY
um), single or precipitation of ANEMIA – most common
multiple small, hemoglobin that has disorder associated w/ Heinz
round, oval or been denatured body. G-6-PDH is an involve
serrated, in hexose monophosphate
purplish shunt. When G6P is
inclusions on RBC converted into 6PG catalyze
periphery by G6PD & the cofactor is
(distorts the NADP which becomes
membrane) NADPH a reducing property
depending on of RBC which in turn
the oxidation maintains GSH into reduced
form, which serves as the
STAINS (SUPRAVITAL major antioxidant property
STAINS): Crystal violet & of RBC which can breakdown
Brilliant Cresyl Blue H2O2 into H2O & O2
preventing oxidation of Hb.
The NADPH formed will
reduced the ferric back to
ferrous thus protecting RBC
to oxidation. Deficiency will
result to deficiency in NADPH
& GSH
 RED CELL INJURY DUE TO
CHEMICAL INSULT OR
UNDER OXIDANT STRESS
(sulfonamides) – causes
accumulation of oxidized Hb
 HEMOGLOBINOPATHIES
AND THALASSEMIA MAJOR
 UNSTABLE HEMOGLOBIN
SYNDROMES
 ZURICH & KOLN HB –
abnormal Hb prone to
precipitation & oxidation,
unstable Hb
 POST-SPLENECTOMY
HB H INCLUSIONS  Small greenish- Precipitate of Hb H, an Seen in Hb H disease
blue inclusion abnormal Hb which has 4 Alpha Thalassemia
bodies polypeptide chains as B
 Seen in occurs when A chain is
unstained cells not completely
 May also be seen synthesize
w/ supravital
stain
 Golf ball w/ pits
when numerous
in RBC
HOWELL-JOLLY BODY  Small, coarse  Nuclear CAUSES:
round granules remnants  SPLENECTOMY
 deep blue or red-  with Feulgen &  FUNCTIONAL
purple, appears Methyl Green HYPOSPLENIA - spleen
singly & is off the Pyronin stains w/ decreased function
center but not at (stains for DNA) thus unable to remove
the periphery  seen when there inclusion
 DNA containing is increase
inclusion karyorrhexis DISORDERS ASSOCIATED W/
meaning it is a (fragmentation KARYORHEXIS:
nuclear fragment of nucleus) •ACCELERATED OR ABNORMAL
 Very tiny ERYTHROPOIESIS
inclusion w/ <1  hemolytic anemias – too
um much RBC are
hemolyzed thus bone
marrow compensate by
accelerating RBC
production thus nuclear
dissolution is also fast &
more nuclear fragments
will be seen
 megaloblastic anemia –
caused by abnormal
nuclear dissolution (Vit.
B12 & Folate deficiency)
PAPPENHEIMER BODIES  small (2-3 um)  Unused Iron  SIDEROBLASTIC
SIDEROTIC GRANULES faint blue deposits ANEMIA – red cell
coccoid bodies precipitate & cannot make use of the
that aggregate in become siderotic iron due to deficiency in
small clusters at granules protoporphyrin 9 thus
the periphery of  Sometimes may iron precipitate &
the cell. appear as Ringed becomes siderotic
 Smaller than H-J sideroblast granules
bodies, deep  THALASSEMIA – affects
blue & are more globin
likely to be  HEMOGLOBINOPATHIES
multiple - affects globin
 Iron containing  ALCOHOLISM – affects
granules positive enzymes in heme
when stained w/ synthesis
Perls Prussian  POST-SPLENECTOMY
blue & named as  CONDITIONS LEADING
siderotic TO
granules & if HEMOCHROMATOSIS or
positive when HEMOSIDEROSIS (iron)
stained w/ – both increase iron or
wrights/giemsa it iron overload
is named as
pappenheimer
bodies
BASOPHILIC STIPPLING  Dark blue Aggregates of ribosomes DEFECTIVE OR ACCELERATED
granules, and mitochondrial HEME SYNTHESIS
multiple, uniform remnants thus acidic in  Lead or other heavy
and evenly component because they metal poisoning –
distributed as took up the basic dye affects hb synthesis
fine dots or large due to rRNA  Thalassemia - globin
granules  Hemoglobinopathies -
 Often confused globin
w/  Megaloblastic anemia -
pappenheimer rarely
bodies (+
Prussian Blue) PYRAMIDINE-5-NUCLEOTIDASE
but basophilic DEFICIENCY
stippling do not Explanation:
stain w/ Prussian When RNA degrades, they
blue thus it is not release their purine &
seen due to lack pyrimidine bases. For the
of iron but if complete degredation of
present in wright pyramidine bases, it needs
& Prussian then Pyramidine-5-nucleotidase thus
it is siderotic deficiency of Pyramidine-5-
granules nucleotidase, RNA degradation
will also be defective or
APPEARANCE: incomplete
COARSE: granules are
much more outlined and
easily distinguished &
larger
PUNCTATE: coalescing
into smaller forms
DIFFUSE: Granules are
fine blue dusting
CABOT RINGS Reddish-violet thread- Remnants of Dyserythropoiesis
like that assumes a loop, microtubules of mitotic megaloblastic anemias
or an incomplete ring or spindle & can reshape Homozygous
a figure of 8 into a loop or a fragment Thalassemias
of the nuclear  Lead poisoning
membrane  Other severe anemia
 Post splenectomy
STRUCTURES NOT SEEN IN WRIGHTS: STRUCTURES NOT SEEN IN WRIGHTS:
1. Granules of reticulocyte 1. Polychromatophilic cell
2. Siderotic granules 2. Pappenheimer bodies
3. Heinz bodies
ABNORMAL RED CELL DISTRIBUTION
AGGLUTINATION Clumping of RBCs Presence of antibodies on  Cold agglutinin
Indication of Ag-Ab the RBC surface produced  Autoimmune
reaction by patient (autoAb) or from Hemolytic Anemia
different person (isoAb)

ROULEAUX  Linear alignment of Increased concentration of  HYPERPROTEINEMIA’S


RBCs appearing as globulin – plasma protein
stack of coins component is altered
 RBC due to resulting to alteration of
destruction of zeta the negative charges
potential surrounding the RBC
 Side by side flat which is seen in
surfaces Multiple myeloma &
 biconcave surfaces Waldenstrom’s
in apposition macroglobulinemia
appearing as a which are plasma cell
stack of coins or discrasias which are
flat plates leukemias affecting
plasma cell causing
increase proliferation of
plasma cells

 MULTIPLE MYELOMA –
most of the affected
plasma cell will
produce excessive IgM

 WALDENSTROM’S

PROTOZOAN INCLUSIONS
 RBC is normal but infected by hematozoic parasite
a. PLASMODIUM SPECIES
• Seen in different stages of smear mostly ringed forms

1) MALARIAL INCLUSION
• Obligate intraerythrocytic parasites
• w/ hepatic stage (liver) where they rapture the liver & proceed to
rupture the RBC & metabolize the Hb but not completely & leave behind a
pigment (hematin or hemozoin pigment)

PIGMENTS INSIDE THE INFECTED RBC:


a. P. vivax - Schuffners dots
b. P. falcifarum – Maurer’s dots
c. P. malariae – ziemann’s dots
d. P. ovale – james dots

2) BABESIA PARASITE
• Parasite of cattle’s causing red fever on cattle’s
• Discovered by babes & may also affect men
• w/ ring forms in RBC
ARTIFACTS
a. FIXATION b. HEAT ARTEFACTS
• When fixative is contaminated w/ water which EFFECTS:
must be water free methanol 1. Red cell bud off into vesicles which might be reported
EFFECTS: microspherocytes
1. Refractile rings in red cells 2. WBC disintegrate
2. Mouth eaten cells – caused by blowing leading to 3. Proteins coagulate producing small weak basophilic
spicules particles which are similar to platelets

WHITE BLOOD CELLS

CLASSIFICATION OF WBC
a. Granulocytes (neutrophil, eosinophil, basophil) & Non-granulocytes (lymphocytes & monocytes)
 Non-granulocytes have granules however too small to be seen
 Cytoplasm of monocytes appear like a ground glass due to the tiny granules present

b. Polymorphonuclears (those w/ segmented or lobulated nuclei – neutrophil, eosinophil & basophil) &
Mononuclears (unsegmented nucleus – monocytes & lymphocytes)
c. Phagocytes (non -specific function – neutrophil (most active phagocyte), monocyte & macrophages,
eosinophil & basophil (both are less efficient phagocytes) & Immunocytes (involve in production of Ab – B
cells)

Formation: Bone marrow & lymphoid tissues (thymus (T-cells), spleen & lymph nodes)
General Function: Defense
NON-MALIGNANT/REACTIVE CHANGES OF WBCS
CYTOPLASMIC MORPHOLOGIC DEFECT OR CHANGE ASSOCIATED CONDITION/S
CHANGES APPEARANCE
TOXIC  Large purple to Altered primary  SEVERE INFECTION – a reaction of the
GRANULATION black azurophilic granules infection
granules especially EXPLANATION: during infection since
in the neutrophils  Neutrophils have neutrophil is involve in this mechanism, it
which contains primary granules undergoes several changes:
primary granules. but when it is 1. EXEMPLIFY & ENHANCE ENZYME
 Larger than 2- altered, it PRODUCTION as they need to
degree granules & appears as a toxic destroy the invading organism by
stain dark blue-black granulation which phagocytosis. The phagocytized
are larger than material must be destroyed thus the
the usual primary cell produces lots of enzymes to
granules perform respiratory burst & must
also alter its granulation by
increasing the function of its
granules. Enzymes such as the
myeloperoxidase is contained in the
primary granules. As the cell
increase its function w/ the
granules, the granules also function
to increase their content resulting to
alteration & toxic granulation.
2. SHIFT TO THE LEFT – even immature
WBC will be released as a
compensation for the severe
infection
3. INCREASED PHAGOCYTIC ACTIVITY
– in infection, the neutrophils have
to phagocytized the invading
organism & the phagocytized cell
will be contained inside the vacuoles
& it will appear as many toxic
vacuolation & granulations. The
cytoplasm also appears as Dohle
bodies as larger inclusion.
 CHEMICAL POISONING
 TOXIC STATES

 Toxic granulation is a normal reaction in


these conditions
DOHLE  Blue, round, or Aggregates of free INFECTION – due to increase activity of the
BODIES/AMATO mostly elongated ribosomes cell
bodies
 Represents rRNA CELLS INCREASE METABOLIC ACTIVITY AS A
 Not permanently RESPONSE IN:
seen in cytoplasm  Severe infections, burns,
except for may- Chemotherapy, surgery
hegglin anomaly  If these conditions disappear,
 Single or multiple Amato bodies will also
arranged in parallel disappear
rows
MAY-HEGGLIN ANOMALY – an inherited
condition, thus appearance of amato is
permanent

 Basophilic stippling: If RNA


precipitates & seen as granules in
RBC
 Amato bodies: if RNA precipitates &
seen as granules seen inside of WBC
 RNA is important in metabolism, as
the cell increases its production of
enzyme, this will increase metabolic
activity thus increased RNA function.
CYTOPLASMIC  Many presence Represents end stage SEPTICEMIA, SEVERE INFECTIONS, TOXIC
VACUOLATION indicate phagocytosis of phagocytosis STATES
particularly the end
stage  During phagocytosis the phagocytize
 Presence of many material will be contained in a vacuole
toxic vacuoles thus increasing the vacuole inside the
containing the neutrophil. Once in the vacuole the
phagocytized phagosome will fused w/ lysosome
material w/ the forming the phagolysosome. Once
contents of lysosome destroyed the debris will be contained in
or debris of the a vacuole
phagocytized
material
 Large empty areas
within the cell
ATYPICAL/  Cells show foamy & Seen in INFECTIOUS
REACTIVE abundant cytoplasm MONONUCLEOSIS/KISSING DISEASE –
LYMPHOCYTE/  ≥40% associated w/ caused by Epstein barr virus
DOWNEY CELLS infectious
mononucleosis

INHERITED DISORDERS
CYTOPLASMIC CHANGES MORPHOLOGIC APPEARANCE ASSOCIATED CONDITION/S
ALDER-REILLY  Large, coarse blue – black granules Associated w/
GRANULATION in all WBCs MUCOPOLYSACCHARIDOSIS/G
 Abnormal polysaccharide granules ARGOYLISM – abnormalities in
which are not completely mucopolysaccharide
metabolize due to an enzyme metabolism
deficiency (congenital disorder) which leads to non-  Hurler’s
metabolism of mucopolysaccharide & prevents the  Hunters
formation of secondary granules

MAY-HEGGLIN  Resembles Dohle bodies but are permanently LEUCOPENIA, VARIABLE


ANOMALY present THROMBOCYTOPENIA, GIANT
 Presence of gray – blue spindle shaped inclusions in PLATELETS
the cytoplasm of Neutrophils and Monocytes

FUNCTIONS OF NEUTROPHIL:
1. DIAPEDESIS – squeezing out through the circulation via the endothelial lining & proceeds via:
Random motility – no particular direction
Direct motility/Chemotaxis – w/ particular direction
2. PHAGOCYTOSIS – killing of phagocytized agent
INHERITED ABNORMAL GRANULOCYTE FUNCTION
CYTOPLASMIC DESCRIPTION
CHANGES
JOB’S SYNDROME  Impaired directional motility
 Random movement of phagocytes is normal thus neutrophil may still move
elsewhere but directional motility is impaired because cells respond slowly to
chemotactic agents thus do not directly proceed to the chemical attractant.
EFFECT: it gives more time for the bacteria to proliferate or cross the infection. If bacteria
are dislodged in the tissue, & immediately phagocytized thus it will not be able to cause
infection but due to slow movement of cells, the bacteria will be able to lodge in the site of
infection causing infection.
LAZY LEUKOCYTE  Both random and directional movement of the cells are defective thus neutrophil
SYNDROME will only stay but the organism will approach the neutrophil thus phagocytosing it
 Cells fail to respond to inflammatory stimuli but have normal phagocytic and
bactericidal activity.

NON-MALIGNANT: DEFECTIVE KILLING OF MICROORGANISMS


CYTOPLASMIC DESCRIPTION
CHANGES
CHEDIAK – HIGASHI 
Characterized by large, blue to grayish round granules or inclusions in Neutrophils
ANOMALY that vary in size and color
 Stain positive with peroxidase, Sudan black B (SBB) and Acid phosphatase (ACP)
stains.
 Represents the fused primary granules. Primary granules contain the usual
composition such as peroxidase, sudan black b & ACP but these are abnormally
package, meaning they are normal in contents but abnormally package, thus
appears as chediak-higashi anomaly.
IN SHORT: granules are normal in content but abnormally packaged thus it cannot perform
its normal function
CHRONIC  Phagocytes ingest all but cannot kill catalase (+) organism because of lack of
GRANULOMATOUS appropriate respiratory burst
DISEASE  Neutrophils have normal direct & random motility & phagocytosis

RESPIRATORY BURST – reaction inside the neutrophil needed for the killing of organism.

In respiratory burst the neutrophil is able to produce potent oxygen radicals like H2O2 &
O2 for killing organisms. In chronic respiratory disease, the respiratory burst function of
neutrophil is decreased or inappropriate thus inadequate production of H2O2 & O2 which
are important bactericidal agent.

Catalase + Organisms: Staph aureus – produce enzyme catalase which will breakdown
H2O2 into H2O & O2

OTHER APPEARANCES
TART CELL  A monocyte (phagocyte) that engulf a lymphocyte (engulf material)
 Formation is no known significance

FERRATA CELL  a stimulated or atypical lymphocyte with denser and more opaque cytoplasm
 associated with SBE
REIDER CELL  a lymphocyte with notched, lobulated, segmented, clover – leaf like nucleus
 associated with Chronic Lymphocytic Leukemia
L.E. CELL (LUPUS  Neutrophil engulf the lysed nucleus of another neutrophil containing a large purple
ERYTHEMATOSUS) homogenous round inclusion (phagocytized cell) with nucleus wrapped around the
phagocytized material which occupies the entire cytoplasm of neutrophil thus
neutrophils nucleus is pushed around the side appearing as a wrapping itself into the
phagocytized cell
 For it to occur there has to be a neutrophil, ANA or L.E. factor & nucleus of another
neutrophil
 Seen in SLE & other autoimmune disorders because it is related to the presence of
anti-nuclear Ab (ANA)
HAIRY CELL  A lymphocyte affected by hairy cell leukemia
 with abundant hair-like cytoplasmic projections

SEZARY CELL  Round lymph cell with nucleus that is grooved or convoluted
 Seen in Sezary syndrome which is an advanced case of mycosis fungoides. Both are T
cell disorder but they represent different stage of the disorder:
Mycosis fungoides – only skin is affected by T cells ; initial stage
Sezary Syndrome – when affected T cell migrate deeper into the dermal layer, lymph
nodes & visceral organs
 Mononuclear cell w/ a brain-like nucleus or cerebriform nucleus & a narrow rim of
cytoplasm
FLAME CELL  plasma cell with red to pink cytoplasm that appears like a flickering candle
 Associated with increase in IgA associated w/ multiple myeloma which is a malignancy
of plasma cell which is responsible for the production of Ig.

GRAPE CELL/ MOTT  plasma cell that contains small colorless (or blue or pink) vacuoles or globules
CELL/ MORULA CELL  large protein globules giving the appearance of grapes
 blue or pink protein that are excessively produced in multiple myeloma

RUSSEL BODIES  accumulation of IgG found in the cytoplasm & also seen in plasma cell

DUTCHER BODIES  intracellular crystalline structures of abnormal IgG


 inclusions of nucleus

NON-MALIGNANT/REACTIVE CHANGES
UCLEAR CHANGES MORPHOLOGIC APPEARANCE DEFECT OR CHANGE ASSOCIATED
CONDITION/S
HYPOLOBULATION 
Neutrophils have single or Decreased segmentation PELGER-HUET ANOMALY:
bilobed nuclei TRUE – inherited
 The 2 lobes appear as a PSEUDO – acquired
peanut or dumbbell shape or
a pince-nez spectacles (pair of
eye glasses)
HYPERSEGMENTATION  Neutrophils show more than 4 Abnormal DNA synthesis Megaloblastic anemia
lobes  Cells are abnormally
 Abnormal in size & lobulation large
& large as a macropolycyte
2 TYPES:
Normal size of neutrophil: 9-15 a. B 12 deficiency
um anemia
b. Folate deficiency
anemia
OTHER NUCLEAR AND CYTOPLASMIC CHANGES
PYKNOTIC CELL  shrunken and dehydrated nucleus of cells that are about to die

PYKNOSIS – cell dissolution

SMUDGE CELL  Nuclear remnant of lymphocyte, seen after smear preparation


 Thumbprint appearance
 Associated w/ Chronic Lymphocytic Leukemia wherein cells are fragile thus they are easily
smudge.

BASKET CELL  Nuclear remnants of granulocytic cell


 Seen in Chronic Granulocytic Anemia
 Basket appearance

BARR BODY  Drumstick like body protrudes away from some of segments of neutrophils & attached on
one of the lobes of neutrophil nucleus
 Indicate an extra X chromosome & seen in females

AUER RODS  Pink or red rod-shaped structures


 Fused primary granules seen in immature neutrophils
 If seen in bundles this are called faggot cells
 Seen in Acute Myeloid Leukemia

INHERITED DISORDERS OF MONOCYTES & MACROPHAGES


Monocytes & macrophages are also involved in phagocytosis & also dispose of. They help in metabolism of lipids &
carbohydrates where they need enzymes & deficiency of these enzymes leads incomplete metabolism thus
incomplete disposal. Non-metabolized substances are contained in the cytoplasm causing macrophage overload.
DISORDER DESCRIPTION ENZYME DEFICIENCIES
MUCOPOLYSACCHARIDOSES  Deficiencies of specific enzymes involved I Hurler’s (most common)
/GARGOYLISM in the degradation of IV Morquio – Ullrich
mucopolysaccharides II Hunter’s
 Non-metabolized mucopolysaccharide will V Scheie
be contained in the cytoplasm of the cell III Sanfilippo
as granules VI Maroteaux – Lamy
 Alder-Reilly granules represents the non-
metabolized mucopolysaccharide. It is
associated w/ the enzyme deficiencies.
LIPIDOSES (abnormal lipid metabolism)
DISORDER DESCRIPTION & ENZYME DEFICIENT
GAUCHER’S DISEASE (MOST  lack of  - glucosidase resulting inability to metabolized glucocerebroside
COMMON) which will accumulate of in the spleen, liver and bone marrow

Gaucher cell – cytoplasm is distended with many glucocerebroside; sponge-like


appearance, crinkled or crumpled tissue paper
NIEMANN – PICK DISEASE  deficiency of sphingomyelinase resulting to accumulation of sphingomyelin
and cholesterol in the macrophages
 Foamy appearance
SEA –BLUE HISTROCYTES  w/ non-metabolized lipid-rich granules that stain blue green with polychrome
stains
 accumulation in the spleen and BM of histocytes giving an appearance of a
blue sea
 s/s: hepatomegaly, thrombocytopenia

FABRY’S DISEASE  lack of a-galactosidase resulting to accumulated glycolipid trihexosyl ceramide


but will not give an unusual appearance of macrophage
WOLMAN DISEASE  deficiency of acid esterase resulting to accumulation of TG and cholesterol
TANGIER’S DISEASE  Patient is unable to produce HDL (good cholesterol) which responsible for the
metabolism of the cholesterol by bringing it into the liver to prevent
hypercholesterolemia, as a result, cholesterol esters accumulate

TESTS FOR NEUTROPHIL FUNCTION: EVALUATION OF RESPIRATORY BURST


NTR (NITROBLUE  screening test for neutrophil function (ability of neutrophils to attack bacteria)
TETRAZOLIUM  test for the ability of normal neutrophil to reduce normal yellow water-soluble dye
REDUCTION) TEST into an insoluble blue formazan
 abnormal neutrophils are unable to reduce the yellow water-soluble dye into blue
formazan
CHEMILUMINESCENCE  Based on the ability of the cell for emission of low-level light pulses by
stimulated cells.
 Normal neutrophils when not stimulated or in resting state do not emit light
but when stimulated they emit a low level of light.
 Abnormal neutrophils w/ impaired respiratory burst cannot emit light whether
stimulated or resting
BOYDEN MICROPORE  Utilizes a micropore filter where neutrophils are counted. This is placed on an
FILTER abrasion to determine the ability of neutrophils to migrate
 Tests migration of neutrophil (fastest to migrate) in response to a chemotactic
factor.
REBUCK SKIN WINDOW  Requires performance of superficial abrasion on the skin to evaluate the
speed, type and number of phagocytes that respond to a skin abrasion.
 On top of the abrasion is the slide. After, perform a count on the slide &
observe.
PLATELET released form BM which may be increased in
 Cytoplasmic fragments of megakaryocyte number in cases of hemolytic anemia &
 Small packages of cytoplasm that are nipped off from hemorrhagic anemia
the cytoplasm of megakaryocyte
 Are complex and metabolically active. They function PLATELET STRUCTURE: FUNCTIONAL PLATELET
in hemostasis by interacting with their environment ZONES
to initiate and control hemostasis
A. PERIPHERAL ZONE: outermost zone
GENERAL CHARACTERISTICS OF THROMBOCYTES  Glycocalyx bears the different glycoproteins (Gp
Composition: 60% proteins, 30% lipids, 8% CHO, 1a, 1b, 1c, 2a, 2b, 3, 4, 5) which are not present
minerals, water, nucleotide, glycogen & glucose (main in other cell membrane. These glycoproteins
ATP source) serves as surface for the adherence of other
Shape & Origin: Thin disc cytoplasmic fragment coagulation factor (CF 1, 5, 7, 11, 12, 13)
Diameter: 2-4 um Ex.: fibrinogen converted into fibrin on the
Volume: 5 – 7 fL surface of platelets
Reference Platelet Count: 150,000 – 450, 000/ uL Platelet can only aggregate to another
Daily Turnover: 35 x 109/L (+/- 4.3) & then destroyed by platelet if it has the important glycoprotein
the spleen & produced by the bone marrow for aggregation
2 pools: 1/3 spleen & 2/3 circulation NOTE:
Lifespan: 8-11 days in vivo SUB-MEMBRANOUS AREA: Underlies the plasma
Function: Hemostasis or stoppage of bleeding by membrane, this contains the communication w/ the
forming into plugs for the maintenance of vascular external environment of cell via receives message
integrity & continuity & blood coagulation from the outside

HEMOSTASIS B. SOL-GEL ZONE:


During vessel injury the blood contained in the  Consists of a stable gel that regulates
vasculature may leave the circulation (extravasation). arrangement of internal organelles.
The blood vessels will constrict to limit the entry of  Contains communication of organelles &
blood into the vessel causing limited blood loss. The maintains location of the different organelles
constriction of vessels is aided by thromboxane A2 &  Where important protein serving as
serotonin from the platelets. Platelets immediately cytoskeleton of the cell is located
form into plugs to prevent further bleeding. The plug is MICROTUBULES (tubulin) & MICROFILAMENTS
an initial plug, w/ the current of flowing plug, the plug (actin & myosin) – provide the cytoskeleton thus
may be removed or dislodge thus bleeding continues. maintaining the discoid shape of platelets
When secondary hemostatic mechanism is activated  When actin & myosin interacts, they form
when fibrin is form of fibrinogen, the fibrin thread will into actomyosin where they form into
be measured around the platelet plug, stabilizing now thrombosthenin
the platelet plug thus further enhancing the hemostatic
effect of platelets. NOTE:
THROMBOSTHENIN – important contractile acting
NOTE: force, provides contractile force after activation of
In the 4 stages of megakaryopoiesis, only the platelets.
metamegakaryocyte (MK 4) with a 4 nucleus or more,
will be able to shed off as platelet. C. ORGANELLE ZONE:
 THROMBOPOIETIN – growth factor for  Where the different organelles, alpha granules
megakaryopoiesis produced mainly by the (most abundant), dense bodies, mitochondria,
liver & some in kidneys lysosomes and peroxisomes are located.
 ENDOMITOSIS – division of nuclear  Alpha & dense granules are for storage of
components but no cellular division thus as important contents while mitochondria for
cell mature it increases in size ATP production, lysosomes containing
 PLATELET BITS/STRING/RIBBON – hydrolytic enzymes
interconnected platelets indicating newly
ALPHA GRANULES CONTENT ROLE IN SUBSTANCE SOURCE
a. High molecular weight kininogen – cofactor in HEMOSTASIS
coagulation system Promote High Molecular Alpha granules
b. Factor 5 – cofactor for coagulation system coagulation Weight
c. Fibrinogen – same as coagulation factor 1, when Kininogen
acted by thrombin it is converted into fibrin thus it Fibrinogen;
is the precursor of fibrin Factor V von
d. Von Willebrand Factor – for platelet adhesion Willebrand
e. Thrombospondin – platelet function Factor
f. Platelet Factor 4 – platelet function Promote ADP; Calcium Dense bodies
g. Beta thromboglobulin – blood vessel repair aggregation Platelet factor 4 Alpha granules
h. Platelet Derived Growth Factor – blood vessel repair Thrombospondin Alpha granules
 When a vessel is injured, it will undergo healing Promote Serotonin Dense bodies
where smooth muscles & fibrous tissues must vasoconstriction Thromboxane A2 Membrane
regenerate. Beta thromboglobulin & PDGF both precursors phospholipids
stimulate smooth muscle repair & chemotactic Promote Platelet derived Alpha granules
to fibroblast vascular repair growth factor
i. Plasminogen – when acted on by its activator, Beta
converts into plasmin ; important for fibrinolysis thromboglobulin
Fibrinolysis – dissolution of the clot by plasminogen Other system Plasminogen Alpha granules
affected Alpha 2-
j. A-2 antiplasmin – Inhibits plasminogen to prevent antiplasmin C1
excessive clot dissolution esterase
k. C1 esterase inhibitor – found in complement system inhibitor

DENSE GRANULES: CAMAS


a. ADP – for platelet aggregation & adhesion
b. ATP – for platelet aggregation & adhesion
c. Calcium – for platelet aggregation & adhesion
d. Magnesium – for platelet aggregation & adhesion
e. Serotonin – for vasoconstriction

PLASMA COAGULATION FACTORS – roman numerals


PLATELET FACTORS – ARABIC NUMBERS
MICROSCOPY FOR PLATELET COUNT
PLATELET FACTORS
 light & phase contrast microscopy
a. PF 3 – platelet phospholipids found on surface of
platelet
PLATELET ESTIMATION
b. PF 4 – inhibitor of heparin thus anti-heparin factor
IMPORTANCE: for correlation for performed platelet
count
MEMBRANE SYSTEM
FORMULA: (total no. of platelets per field) / 10 x 20, 000
 2 membrane systems REPORTING: qualitative/descriptive reporting
a. OPEN CANALICULAR SYSTEM – delivery routes thus Ex.: (70 / 10) x 20,000 = 140, 000 ; slight decreased
it is where nutrients enter & waste products leave  Correlate w/ the chart
the cell
b. DENSE TUBULAR SYSTEM – remnant of endoplasmic
TYPE OF BLOOD SAMPLE PERFORMED
reticulum ; site of arachidonic acid synthesis and
EDTA
functions as a calcium sequestering pump
 because it prevents platelets from adhering &
aggregating together thus appear separated
under the microscope but if examined via fresh
blood, platelet appears clump or aggregated
 DISADVATAGE OF EDTA: platelet sateletism for QUALITATIVE PLATELET DISORDERS
patients who develop Ab against EDTA ADHESION DEFECTS:
1. BERNARD-SOULIER: DEFICIENCY/DEFECT IN GP IB-IX
DIRECT PLATELET COUNT REPORTING: 150 – 450 x 109/L  Characterized by large platelets,
thrombocytopenioa, prolonged and Bleeding
PROPERTIES AND FUNCTIONS OF PLATELETS Time.
1. ADHESION: platelet to non-platelet  Platelets show normal aggregation with
 Adherence to injured blood vessel epinephrine, ADP and collagen but not with
 Ability of platelets to attach/stick to non- ristocetin.
platelet surface. This allows platelets to adhere
to damaged endothelium. 2. VON WILLEBRAND’S DISEASE
 requires plasma von willebrand factor and gp Aggregation test: same with Bernard-Soulier
ib-ix
PLATELET AGGREGATION STUDY TEST
NOTE: MODE OF ACTION  Platelets + agonist (aggregating agents) =
When activated or discoid, they change their stimulate aggregation
morphology where their cytoplasm will have RESULT: BOTH AGGREGATES TO: ADP, collagen &
expansions for adherence. When an injury occurs, epinephrine
some of the connective tissue & smooth muscles BOTH DO NOT AGGREGATES TO: Ristocetin
components enters the circulation & be exposed to
the circulation such as the collagen. Collagen is AGGREGATION DEFECTS
outside the blood vessel which maintains the integrity
of blood vessels, but during injury, some of the 1. GLANZMANN’S THROMBASTHENIA:
collagen are exposed to the plasma & platelets. The DEFICIENCY/DEFECT IN GP IIB-IIIA
platelets adhere on the exposed collagen via the GP  Platelets show normal aggregation with
1B9 complex but GP 1B9 complex will not adhere ristocetin but not with epinephrine, ADP nor
directly to the collagen but needs a receptor the, von collagen.
willebrand factor thus allowing adhesion to occur. 2. AFIBRINOGENEMIA (severe/absent),
HYPOFIBRINOGENEMIA (decrease) OR
2. PLATELET RELEASE REACTION DYSFIBRINOGENEMIA (defective)
 Alpha & dense granules release substances that
will contribute to platelet aggregation and RELEASE DISORDER/GRANULE DEFECTS/STORAGE
activation of the coagulation system GRANULE DEFECT
Ex.: Serotonin for vasoconstriction – when
vessels are injured vessels constrict 1. Gray platelet syndrome – deficiency in Alpha
Thromboxane A2 form the cytoplasm of the granules where platelets appear large pale or gray
membrane phospholipids or pale blue wherein platelet must be blue or purple
ADP from the dense granules promotes 2. Wiskott-aldrich syndrome – deficiency in dense
aggregation for efficient plug strength granules that appears as smallest platelets
3. Chediak-higashi – deficiency in dense granules
3. PLATELET AGGREGATION: platelet to platelet 4. Hermansky-pudlak syndrome – deficiency in dense
 Platelets stick to one another platelet to form granules
an initial platelet plug.
 Induced by stimuli such as ADP, thrombin, TxA2, QUANTITATIVE PLATELET DISORDERS
collagen & epinephrine.
 Requires Gp IIb-IIIa and plasma THROMBOCYTOPENIA: (<150, 000)
fibrinogen/FACTOR 1. 1. Decreased production – BM is ineffective in
 FACTOR 1 serves as the adhesive glycan or producing platelets due to:
glue of the 2 platelets together a. Megakaryocyte Hypoproliferation
b. Ineffective thrombopoiesis
4. VASOCONSTRICTION & CLOT FORMATION
c. Marrow replacement/myelopthesis – BM space a. Uremia – increase in NPN thus damaging cells
is replaced by abnormal cells (cancer, leukemic b. Paraproteinemias - increase in abnormal
or myeloma cells) protein
THROMBOCYTOSIS/THROMBOCYTHEMIA
2. Dilutional – related to massive blood transfusion
which is the transfusion of >1.5 L blood per 24 hr. 1. Primary Thrombocytosis – excessive production is
The available circulating platelet of the patient will the main defect
be diluted by transfused blood. If transfused blood  Myeloproliferative diseases – disorder of the
is not fresh do not have viable platelets. bone marrow that results to excessive
 Outside the body platelets lifespan is only 3 production of blood cells such as in cases of
days polycythemia vera which is excessive
production of all blood cells ; uncontrolled
3. Increased destruction (in vivo) production caused by the permanent damage of
a. Immune – presence of Ab mostly IgG such as in bone marrow
immune thrombocytopenic purpura  Essential Thrombocytosis – excessive
ITP – caused by autoimmune Ab destroying the production of abnormal platelet
platelets
2. Secondary/Reactive thrombocytosis – reaction to
b. non-immune – no Ab implicated such as in another condition ; irreversible
abnormal activation of platelets (thrombotic  hemolytic anemias
thrombocytopenic purpura)  hemorrhagic blood loss anemia
TTP – platelets are consumed as they are
converted into platelet aggregates or thrombus BOTH: body will compensate for the loss of
leading to decreased in platelet count blood by causing splenic mobilization wherein
the 1/3 in the spleen will move into the
4. Splenic sequestration – in cases of splenomegaly, circulation as a compensation thus transiently
platelets will be sequestered more in spleen than in increasing platelets but returns to normal after
circulation thus decreased platelet count a few hours
 splenectomy
5. Acquired
SPECIAL HEMATOLOGY PROCEDURES for 2 hours until 3rd hour. Perform interval
TEST FOR SICKLING HEMOGLOBINS microscopic examination.
 POSITIVE RESULT: 1. Sickle RBC (Hb S) but does
 SICKLE CELLS are distorted cells appearing as determine if heterozygous AS or homozygous SS
thin, dense and elongated cells with both ends
 REPORTING: positive or negative
pointed. SICKLING is caused by the
PRECIPITATION of an ATYPICAL HEMOGLOBIN NOTE:
(commonly hemoglobin S) which DISTORTS the  Degree of sickling depends on the
RED BLOOD CELLS into a SICKLE or CRESCENT concentration of the Hb S in the RBC
SHAPE. HEMOGLOBIN S is fully soluble when  Readings are made at an hourly interval for 2-
oxygenated but becomes insoluble when the 3 hours
oxygen level is decreased. It first POLYMERIZES  POSITIVE RESULT is diagnostic but does not
then forms into CRYSTALS which cause the red distinguish hb s trait & hb s anemia
cell to become RIGID.

SCREENING TESTS 2. SODIUM METABISULFATE METHODS (DALAND


& CASTLE)
 PRINCIPLE: HbS forms an insoluble tactoid  REAGENT: 2% Sodium metabisulfide or sodium
crystals when oxygen supply to the red cell is bisulfate which are reducing agent which bind &
decreased. When Hb S is deoxygenized outside remove O2 from the blood
the cell into the plasma it will cause the solution  PRINCIPLE: a drop of blood is mixed w/ a drop
to be TURBID. of 2% sodium metabisulfite (a reducing agent)
 Positive result: on a slide, & the mixture is sealed under a
o Sickle RBC coverslip. The hemoglobin inside the RBCs
o Turbidity becomes deoxygenated causing polymerization
 Degree of sicking or turbidity depends on the & the resultant sickle cell formation
concentrstion of HbS in the red cell is decreased.  PROCESS: add a drops of blood & the reagent
HB S in the RBC into the center of the slide. Cover the slide w/
coverslip & seal the sides. Observe
1. SEALED WHOLE BLOOD METHOD (SCRIVER & microscopically.
WAUGH)  POSITIVE RESULT: sickled RBC deoxygenated by
 PRINCIPLE: RBC take on a SICKLE-LIKE SHAPE reducing agent
when oxygen supply to the red cell is decreased.
HbS forms INSOLUBLE TACTOID CRYSTALS when 3. DITHIONITE SOLUBILITY TUBE TEST
exposed to LOW OXYGEN TENSION.  REAGENT: sodium hydrosulfite (dithionite)
 PROCESS: Perform IN VIVO. Using a RUBBER removes O2
BAND, the rubber band is TIED at the BASE of the  SPECIMEN: whole blood w/ EDTA heparin or
FINGER or at the BALL of the finger intended to sodium citrate in order to prevent clotting
be prick & maintain it for at least 5-10 minutes  PRINCIPLE: Red cells are lysed by saponin
to cause obstruction in order to obstruct the allowing Hb to escape. Sodium dithionine binds
circulation thus decreasing the oxygen & Hb S & removes oxygen from the test environment.
will form into crystals. Prick the deoxygenated Hb S polymerizes in the deoxygenated state &
finger & drop into the center of the slide & cover forms a precipitate in a high-molarity
w/ cover slip. Seal the sides w/ petroleum jelly or phosphate buffer solution. The tactoid refract
paraffin to prevent entry of atmospheric O2 or deflect light & make the solution turbid.
which contains abundant O2 which may cause  PROCESS: anticoagulated whole blood + sodium
the sickled cell to revert to normal shape. dithionite. Saponin lyses the RBC releasing now
Incubate for 1 hour at room temperature & Hb. Hb binds w/ sodium dithionite which
examine microscopically if still none, incubate removes O2 from solution causing Hb to
crystalizes but polymerization happens outside
the RBC thus it will not cause sickling but RETICULOCYTE COUNT AND ITS ASSOCIATED
turbidity of solution & is determined by CORRECTIONS
refraction & deflection of the solution in the RETICULOCYTES
presence of light. Observe specimen for
 Are young erythrocytes that are in a discrete,
turbidity by holding the tube 2.5 cm in front of a
penultimate phase of maturation where the
newsprint or a card reader w/ thin black line
nucleus has been removed, however, some of
 RULE: if the solution is observed in the solution
the extranuclear RNA remains in the cytoplasm.
then the solution is not turbid thus negative
 The term “reticulocyte” is derived from the fact
result but if lines are no longer visible then it is
that the cell contains a small network of
positive for tactoid crystals & sickling.
basophilic materials called reticulum which is
 POSITIVE RESULT: turbidity
demonstrable only by supravital stain.
NOTE: Reticulocytes is the 5th stage of erythropoiesis
 Turbidity indicates presence of sickling Hb having no nucleus as well as contains remnants
regardless of any genotype of RNA. The presence of RNA is used to
 Screening method of choice determine presence of reticulocyte which is
stained w/ supravital stain which will precipitate
the RNA into granulofilamentous particles.
4. HEMOCARD Hb A & S
 In the peripheral blood using Romanowsky stain,
 PRINCIPLE: Hb A & S contains monoclonal Ab
they are called as polychromatophilic
(IgG), which specifically bind to the amino acids
erythrocytes. Reticulocyte count and its
at or near the 6th position of the B-chain of the
associated corrections can be used to assess
Hb S & A
bone marrow erythrophoietic activity. Wherein
NOTE: BM is the main organ that produces
 Hb S monoclonal Ab will react w/ the B-chain erythrocytes.
of Hb S but not w/ the Hb A  PRINCIPLE: The ribosomal RNA is stained
 Hb A monoclonal Ab will react w/ the B-chain supravitally. Any non-nucleated RBC that
of the Hb A but not w/ Hb S contains 2 or more blue-stained
 Abs are adsorbed to suspended metal sol particles/granulofilamentous materials, is
particles giving the reagent raspberry-like counted as reticulocyte.
color
 PROCEDURE: In a test tube, add an equal
number of blood with supravital stain (new
NOTE: methylene blue or brilliant cresyl blue or janus
 After electrophoresis, densitometry is green for granulofilamentous staining). Incubate
performed to determine the density of the for 10-20 minutes inside of an incubator to
bands created by the migrating proteins enhance the staining because staining of living
converted into concentration. cell is longer because they resist staining due to
 In cellulose Acetate, at an alkaline pH, Hbs are their intact membrane. After incubation, remix
negatively charge thus they migrate towards the sample because the reticulocyte has a low
the anode. Hb S together w/ D & G upon specific gravity thus they tend to float in the
migration appears as a band of protein thus
solution. After, transfer a drop into a slide &
densitometry is performed to determine the
prepare smear.
concentration of Hb S.
 On Citrate Agar, at an acidic pH, Hb S migrates  METHODS OF STAINING: Wet method and Dry
towards the anode thus citrate agar is more Method (schilling’s method)
confirmatory. METHOD OF COUNTING:

A. LIGHT MICROSCOPE: DIRECT COUNT


 Reticulocytes are enumerated among 1000
RBCs in areas where RBCs are close but not
overlapping and reticulocytes appear to be well out of focus appear as refractile, while
stained. granulofilamentous particles will disappear.
 Formula:
ASSOCIATED CORRECTION
RETIC % = (No. of Retics / 1000 RBCs observed) x 100 ABSOLUTE RETICULOCYTE COUNT

B. MILLER DISC METHOD  Actual no. of reticulocytes in 1 L of whole blood


 Miller Disc is a small disc, connected to one of o ARC = Retic% / 100 x RBC count =
the ocular (_x1012/L) x 1000
 The calibrated Miller disc appears in the field o EXAMPLE: (1.5% / 100) x 4 000 000 = 60,
with 2 squares: a large square and a small square 000
inside the large square which is 1 /9 the size of o REFERENCE VALUE: 25, 000 – 75, 000
the larger square which can be located anywhere
CORRECTED RETIC COUNT / RETICULOCYTE INDEX
 Reticulocytes are counted in the large square
while RBCs are counted in the small square in  This corrects the reticulocyte count to a normal
successive fields on the film until a total of 500 Hct to allow correction for the degree of patient
RBCs have been counted anemia. The percentage of reticulocytes may
 RULE: include in the count the reticulocyte appear increased because of early release into
present in small squares the circulation or because of a decrease in the
no. of mature RBC in the circulation
FORMULA:
𝒕𝒐𝒕𝒂𝒍 𝒏𝒐. 𝒐𝒇 𝒓𝒆𝒕𝒊𝒄𝒔 𝒊𝒏 𝒕𝒉𝒆 𝒍𝒂𝒓𝒈𝒆 𝒔𝒒𝒖𝒂𝒓𝒆
o CRC: Retic % x (Hct (L/L) / 0.45 L/L)
Retic (%)= × 𝟏𝟎𝟎 0.45 L/L = normal mean Hct
𝒕𝒐𝒕𝒂𝒍 𝒏𝒐.𝒐𝒇 𝑹𝑩𝑪𝒔 𝒊𝒏 𝒕𝒉𝒆 𝒔𝒎𝒂𝒍𝒍 𝒔𝒒𝒖𝒂𝒓𝒆 ×𝟗
REFERENCE VALUE: o REFERENCE VALUE: 1% (depends on the
ADULTS: 0.5 - 1.5 % degree of anemia)
NEWBORN: 2.5 - 6.5 %
RETICULOCYTE PRODUCTION INDEX/SHIFT
CORRECTION (RPI)
NOTE:
 More appropriate test for determining efficiency
 Counterstain (wrights or giemsa) is not
advisable because it may destroy the image of BM compensation for anemia
 Reticulocyte have lower specific gravity  General indicator of the rate of erythrocyte
 Increased glucose inhibits staining thus production increase above normal in anemias
granules may not be seen when observed. meaning the previous production rate increases
for compensation for blood loss
 Index calculated to correct for the presence of
INCLUSIONS CONFUSED WITH RETICULOCYTES
shift reticulocytes that otherwise may falsely
 Pappenheimer bodies – iron containing elevate the visual reticulocyte count.
granules ; stain w/ Romanowsky or Prussian
 Done if shift cells/stress reticulocyte are present
blue stain (siderotic granules)
& computing it
 Howell-Jolly bodies – appears singly ; stain
 RULE: the more severe the anemia, the higher
smear w/ Romanowsky to differentiate
the production must be to normalize the
 Heinz bodies – appears pitted golf ball ; stain condition
w/ supravital stain o RPI = Corrected Reticulocyte count /
 Hb H – appears at the periphery attached to Maturation time (days)
the membrane of RBC ; stain w/ supravital Maturation Time: depends on the
stain patient’s hct
 Artifact – if stained smear is stored longer it o REFERENCE VALUE: 1 maturation time if
may create into precipitate which may hct is 0.45
deposit in the surface of RBC & be mistaken
as the granules thus filter the stain & move
the fine adjustment. Artefacts when you are
RETICULOCYTE MATURATION spherocytic cell and functional state of the cell
HCT TIME (DAYS) membrane.
40-45% 1.0  SPECIMEN: Heparinized blood or defibrinated
35-39% 1.5 blood because other anticoagulants alter the pH
25-34% 2.0 of the blood
15-24% 2.5 o Blood sample should be obtained with
<15% 3.0 minimum trauma and stasis thus upon
NOTE: collection of the blood there must be no
 Reticulocyte mature in circulation after 1 day hemolysis
but in the presence of anemia the bone o Ensure uniform size of blood drops
marrow has to release even the immature
reticulocyte. Immature reticulocyte stay NORMAL RESULT:
longer in the circulation a. Incomplete hemolysis: 0.45 % saline, solution
 RULE: the shorter the immature reticulocyte w/ pinkish tinge & an intact button of RBC at
spent in the bone marrow, the longer they are the bottom upon centrifugation
in the circulation b. Complete hemolysis: 0.35 % saline, red
 RULE: the lower the Hct, the longer the solution w/out an intact button at the bottom
maturation time because of the severity of
anemia Note: Spherocytes show hemolysis as early as 0.60%
saline indicating prone to lysis or fragility
CLINICAL SIGNIFICANCE OF RETICULOCYTES
METHODS:
 Increased in hemolytic & hemorrhagic anemias
1. SANFORD METHOD
OSMOTIC FRAGILITY TEST (OFT)
 Whole blood is pipetted to each of a series of
 OFT measures the ability of the RBCs to take in
hypotonic saline solutions of graduated
fluid without lysing. As the cells take in water
concentration.
(hypotonic solution), they become bloated or
 Visual observation
swollen. If the cell is normal or biconcave disk, it
 (+) result: Hemolysis
can take in more water w/out lysis because the
biconcavity can be distended w/out lysing but
2. DACIE’S METHOD
the cell is already a spherocyte, its volume is
 Uses hypotonic saline buffered with PO4 at ph
occupied thus as it takes more water, it will
7.4 Hemolysis is determined
immediately hemolyzed. It Reflects RBC shape
spectrophotometrically at 540 nm
and size (surface area to-volume ratio). This is
 If the degree of spherocytosis is mild the degree
used to detect spherocytosis, because
of hemolysis is not detected at first thus incubate
spherocytes rupture in saline concentrations
for hours to determine presence of hemolysis
near the normal level. It may also detect target
 (+) result: Hemolysis
cells, which, owing to their reduced hemoglobin
content, are able to withstand osmotic stress NOTE:
and rupture only at very dilute saline  INCUBATED OFT: sterile incubation at 37C for
concentrations. 18 – 24 hrs to detect mild forms of hereditary
 IMPORTANCE: Diagnosis of Hereditary spherocytosis.
Spherocytosis (HS) and other hemolytic anemias
associated with spherocytes.
CLINICAL SIGNIFICANCE OF OFT
 ABILITY OF CELL W/OUT LYSING IS AFFECTED
BY: shape & size of the RBC, which in turns a. INCREASED OFT (cells are fragile): Hereditary
depends on the volume, surface area wherein a spherocytosis; red cells with abnormal
hypochromic cell has a wider area than membrane; severe G-6-PDH deficiency;
Pyruvate kinase deficiency; hemolytic anemias
characterized by abnormal membrane or 3. FINAL SETTLING/PACKING
enzyme deficiency  occurs during the last 10 minutes settling slower
b. DECREASED OFT (cells are resistant to lysis due at the bottom of the tube
to enough surface area to volume ratio thus
FACTORS AFFECTING SETTLING OF RBC
these are hypochromic cells & target cell): Sickle
cell anemia, severe Iron deficiency anemia, 1. ERYTHROCYTE FACTORS
thalassemia  RBC SIZE OR MASS: directly proportional to ESR
o Sickling is reversible, if a sickle cell or rate of settling
intakes water it reverses back to its  RULE: the larger, the faster is the settling & the
normal shape, thus ability of sickle cell smaller RBC is, the slower they settle
to react in a hypotonic solution will  NUMBER OF RBC: inversely proportional
depend on the degree of sickling o In anemia where there are only few
ERYTHROCYTE SEDIMENTATION RATE cells present thus more space to settle.
In anemia, lesser cells, faster settling. In
 Measures the degree or rate of settling of
polycythemia vera, cells are crowded
erythrocytes at the bottom of the tube in
thus settling is slower.
plasma in an anticoagulated whole-blood during
a specified period of time (1 hour)  PRESENCE OF ANISOCYTES & POIKILOCYTES:
slower settling due to no rouleaux formation
IMPORTANCE: wherein these cells are abnormal in shape &
size
1. Non-specific measure of inflammation
o The main factor affecting the rate of
2. PLASMA FACTORS
settling is the presence of varied plasma
 PLASMA VISCOSITY: inversely proportional to
protein such as acute phase proteins
ESR meaning the more viscous the plasma is, the
produced during inflammation
slower is the rate of settling
2. It is a good index for the presence of a hidden but
o The most important factor which affects
active disease like in the case of inflammation &
the rate of settling is the plasma
tuberculosis
composition
3. A hematology test to indicate inflammatory
response to tissue injury however it does not  PLASMA COMPOSITION: increase fibrinogen, α-
determine the severity of inflammation 1 globulin, α-2 globulin: increase ESR thus
4. Follow the progress of certain disease but not altering the zeta potential of cells causing
identify the severity of the condition formation rouleaux
5. It measures the suspension ability of the RBCs o increase albumin & lecitihin: decreases
6. It measures the abnormal concentration of ESR due to rouleaux formation inhibition
fibrinogen and globulin
3. MECHANICAL/ TECHNICAL FACTORS
STAGES OF ESR:  POSITION OF THE TUBE: should be exactly
straight vertical because a slight tilting of the
1. LAG PHASE/INITIAL ROULEAUX
tube 3 degree causes 30% error thus faster
 occurs during the first 10 minutes & the settling
settling
is slower but after the first 10 minutes where
 INCUBATION ENVIRONMENT: should be free
they form in rouleau, RBC sediment faster
from any movement or vibration at room
 ROULEAU – cells in apposition to one another w/
temperature however fluctuation caused
their flat surface
vibration causes a significant error because
vibration causes more cells settle at the bottom
2. DECANTATION PHASE/ PERIOD OF
 INCUBATION TEMPERATURE: should be at room
RAPID/CONSTANT SETTLING
temperature
 occurs within 40 minutes.
 LENGTH AND DIAMETER OF THE TUBE: the
longer & wider the tube, the faster is the ESR due
to increase pressure & wider space for cells to ESR: WESTERGREN METHOD
settle down AGE MALES FEMALES
 ANTICOAGULANT: should not be over- <50 y/o 0-15 mm/hr 0-20 mm/hr
anticoagulated (EDTA or citrate) of causes slower >50 y/0 0-20 mm/hr 0-30 mm/hr
ESR because cells are shrunken which may not >85 y/o 0-30 mm/hr 0-42 mm/hr
form into rouleau Children: 0-10 mm/hr
 TIME OF SETTING UP: first 2 hours or 6th hour CLINICAL SIGNIFICANCE OF ESR
from collection because beyond that cells will a. INCREASED ESR IN DUE TO INFLAMMATION OF
swell thus no rouleaux formation THE TISSUES
 Inflammations; acute & chronic infections,
ESR METHODS tuberculosis, multiple myeloma
1. WINTROBE & LANDSBERG METHOD  Rheumatic fever, rheumatoid arthritis,
(WINTROBE TUBE) myocardial infarction, nephrosis
 WINTROBE TUBE: glass tube w/ thick walls  SBE, Waldenstrom’s macroglobulinemia,
where one end is open & the other is close & hepatitis, menstruation, pregnancy
feeding of blood is done manually, w/ 2
graduation 10 on top & 0 at the bottom (left) b. DECREASED OR NORMAL ESR IN
for microhematocrit reading while for ESR it is 0  Polycythemia due to many blood cells,
on top & 10 on the bottom (right). Internal spherocytosis, conditions associated with Hb S
diameter is 3 mm & total height is 11 cm but & Hb C disease
column of blood is 10 cm  TO DETERMINE DECREASED ESR: if cells do not
 MATERIAL: wintrobe tube, Pasteur pipet w/ settle at all
thin needle pipet
 ANTICOAGULANT: Ammonium-Potassium OTHER METHODS OF ESR
Oxalate or wintrobe mixture or double oxalate 1. GRAPHIC-CUTLER METHOD: uses 3.8% sodium
citrate and Cutler tube
2. WESTERGREN METHOD (WESTERGREN TUBE) 2. LINZENMEIER METHOD: uses 3.8% sodium
 WESTERGREN TUBE: plastic or glass tube, both citrate and Linzenmeier tube
ends are open. Length is 200 while the blood 3. MICRO-LANDAU METHOD: uses 5 % sodium
column is 200 mm. The internal diameter is 2.65 citrate
(+- .15) 4. SMITH MICRO METHOD: uses 5 % sodium citrate
 MATERIALS: Westergren tube w/ stopper, 5. CRISTA OR HELLIGE-VOLLMER METHOD
commercial tube w/ anticoagulant, Westergren 6. ROARKE-ERNSTENE METHOD: uses heparin
rack 7. BRAY’S METHOD: 1.3% sodium oxalate
 ANTICOGULANT: Original Westergren = 3.8% NEW ESR METHOD
sodium citrate 1. Automated Erythrocyte Sedimentation Rate
 Modified Method = 2 ml of EDTA- a. Ves-Matic system – based on optoelectronic
anticoagulated blood with 0.5 ml NSS or 3.8% sensor wherein it measures the change in
sodium citra as diluents in order to adjust hct opacity of a column of blood as
 REFERENCE: adjust hct 0.35 sedimentation occurs
 PROCESS: Westergren tube is pushed into a b. Sedimat 15 – based on infrared
bottle of blood, the blood will move up via measurement wherein tubes are slightly
capillary action making sure that the blood is inclined thus the normal values are also
properly mixed via inversion & incubate for 1 adjusted
hour in ESR rack by letting it stand on a c. ESR STAT PLUS system - based on how fast
Westergren rack cells settle upon centrifugation for pediatric
 REPORTING: mm/hr or mm patients because it requires smaller samples
o More accurate if hct is adjusted to 0.35
to limit effect of anemia on RBC
ZETA SEDIMENTATION RATIO (ZSR) NOTE:
- Uses zetafuge which applies controlled ABSOLUTE EOSINOPHIL COUNT:
centrifugation of blood, producing alternating  FORMULA: Absolute count x WBC count
compaction and dispersion of erythrocytes to WBC COUNT: 9000/ uL
measure how closely the erythrocytes approach DIFFERENTIAL COUNT:
one another under a specific standardized  NETROPHIL: 0.60
gravitational force  LEUKOCYTE: 0.30
- Also uses small amount of blood due to a special  MONOCYTE: 0.08
 EOSINOPHIL: 0.02
capillary tube, 75 mm in length with an internal
NOTE:
diameter of 2 mm
 There are some cells that needs to be counted
- After centrifugation, Zetacrit% is determined at
directly such as the eosinophil & basophil
the “knee” of curve to determine zeta  We rarely see eosinophil & very rare for
sedimentation ratio basophil thus performing count directly &
manually is necessary
FORMULA:
𝒉𝒄𝒕 %
ZSR (%) = × 𝟏𝟎𝟎
𝒁𝒆𝒕𝒂𝒄𝒓𝒊𝒕 %
THORN TEST (Test for adrenocortical functions)
REFERENCE VALUE:
(ZSR): 41 – 54% - Does not determine bone marrow function but
adrenal cortical function
- Perform direct absolute eosinophil count w/
ZETA SEDIMENTATION RATIO fasting sample w/ ACTH injection. After,
51-54% Borderline normal introduce ACTH in order to stimulate the adrenal
55-59% Mildly elevated gland to produce corticosteroid hormones which
60-64% Moderately elevated will cause the circulation to leave the circulation
>65% Markedly elevated & proceed towards the tissues. After 4 hrs., draw
second specimen for absolute eosinophil count.
ADVANTAGES OF ZSR After 4-8 hours ACTH injection, when the adrenal
gland reacts it will produce corticosteroid
 It requires a smaller amount of blood hormones causing now eosinophil to proceed to
 It is not affected by anemia and is faster than the tissues thus giving a lower count. If the
other methods adrenal gland function is normal it will be able to
 Reference range is the same for both sexes cause increase production of adrenal hormones.
 Has only 1 reference value - NORMAL: 2nd count should at least be 50%
 Faster due to involvement of centrifugation lower than initial count
o If 1st & 2nd count is the same meaning
ABSOLUTE EOSINOPHIL and BASOPHIL COUNTS
adrenal gland is not functioning causing
 DILUTION FOR EOSINOPHIL & BASOPHIL: 1:10 hypoadrenalism
to increase observation because the higher the
dilution the fewer the cells seen ABSOLUTE BASOPHIL COUNT
 DILUTING FLUID FOR EOSINOPHIL GRANULES:
- uses Cooper and Cruickshank stain (neutral red)
o Phloxine – stains eosinophil red
- COUNTING CHAMBER: Speirs – Levy or Fuchs-
o Propylene glycol – lyses RBC
Rosenthal
o Heparin – inhibits leukocyte clumping
- DILUTION: 1:10
o Na Carbonate – enhances eosinophil
granule staining & hemolyze other WBC LUPUS ERYTHEMATOSUS (L.E.) CELL PREPARATION
o ALTERNATIVE STAINS: Pilot’s solution & - An LE cell is a neutrophil or macrophage that has
Randolph’s stain phagocytized (engulfed) the denatured nuclear
material of another cell that has been lysed by
anti-nuclear Ab. Its formation is characterized by
the production of a number of autoantibodies. - serological test
The most important of these are the antinuclear - Uses fluorescein-conjugated antihuman IgG
antibodies which occur in the serum of patients
BONE MARROW STUDY
with some autoimmune disorders including
COLLECTION OF SPECIMEN
systemic lupus erythematosus (SLE).
Demonstration of LE cells therefore suggests the  ADULTS: posterior iliac crest (posterior
presence of these antinuclear antibodies also preferred or anterior) – more common because
termed as the LE factor the tissue is more thinner at the hips & sternum
TO PERFORM LE:  CHILDREN: tibia

 The LE factor or antinuclear antibodies causes PROCESS: the aspirating needle must be able to reach
nuclear lysis (lysed nucleus becomes a the medulla
homogenous amorphous mass) and this material  Examination of BM requires examination of
is then phagocytized by a neutrophil. blood film which must be collected first because
 Extruded nuclei is acted by antinuclear bone marrow collection requires boring
antibodies because the outer layer of bone is calcified thus
 The phagocytic neutrophil which has it is very painful & stressful for the patient
phagocytized the lysed nucleus is now called an which causes falsely increase WBC
L.E. cell. (seen on a buffy coat smear)
 Rosette formation a flower like formation which 1. TREPHINE BIOPSY/CORE
has an extruded nucleus surrounded by smaller - Utilizes the jamshidi needle for obtaining the
neutrophils is NOT considered positive core to get a rounded portion of the BM tissue
thus it will not disturb the nearby tissue
PROCEDURE:
- This is taken before aspiration biopsy to avoid
a. Collect blood w/out an anticoagulant, 10-15 mL any disruption of marrow architecture.
WB & allowed to completely clot for 1 hr at 37 C - Requires small sample of BM
or 2 hr at RT - Imprint biopsy is prepared by touching the
b. Discard the serum to demonstrate the LE cell by specimen on a slide.
obtaining the buffy coat layer - FIXATIVE: 5% Zenker fluid
c. Get the blood clot & macerate to extrude some
of nucleus thus if antinuclear antibodies is 2. ASPIRATION
present it will act on the extruded nucleus thus - This follows immediately trephine biopsy
lysing the nucleus - Disturb the architectural pattern of cell
d. Macerated blood will be transferred into 3-4 - 1-3 mL of marrow in a 30-35 mL syringe w/ EDTa
wintrobe tube & centrifuge at 2550 rpm for 30 to prevent cellular distortion & processed within
mins. 1hr.
e. After centrifugation, there is a higher volume of - NEEDLE: university of Illinois sternal needle
buffy coat
BONE MARROW SMEAR PREPARATION:
f. Remove serum to obtain buffy coat & perform
buffy coat smear 1. WEDGE OR COVERSLIP METHOD – 2 coverslip
g. Stain w/ wirghts & giemsa & observed for LE cell method giving excellent distribution of blood
which is a neutrophil that has engulf a large mass cells
h. Phagocytic neutrophil appears to wrap into the 2. PARTICLE SMEAR/SQUASH METHOD – on slide
lysed nucleus place the core collected & cover w/ another slide
& squash
OTHER ANTI-NUCLEAR ANTIBODY TEST
3. BUFFY COAT (CONCENTRATE) SMEAR – collect
a. FLUORESCENT TEST (has replaced the LE test)
in a wintrobe tube & prepare buffy coat smear
- more accurate because it identify what kind of
same w/ wedge smear & centrifuged for 8-10
nuclear Ab is produced by patient
mins. at 2800 rpm this will compensates for
hypocellular marrow and allows for examination o HYPOCELLULAR/HYPOPLASIA:
of large numbers of nucleated cells without incomplete development in one or more
interference from fat or RBCs. lines thus it contains more fats thus less
4. HISTOLOGIC SECTION (CELL BLOCK) blood production like in aplastic anemia
o FIXATIVE: 10% formalin, Zenker glacial
MARROW DIFFERENTIAL
acetic acid, or
- include all nucleated hematopoietic cells EXCEPT
o B5 FIXATIVE STAINS: Romanowsky; iron
megakaryocytes & macrophages
stain; H and E
- at least 500 cells are counted preferably 1000
NORMAL MARROW CELLS cells, 500/slide (2 slides)

 Hematopoietic cells – stain w/ romanowsky Guidelines for adult bone marrow differential in
stain concentrated smears, 1000 cell counts
 Lymphoblasts CELL TYPE RANGE (%)
 Monocytic cells Eryhroblast 18-24
 Myelocytic cell Myeloblasts, Type I 0-1
 Erythrocytic precursors Myeloblasts, Type II 0-2
Promyelocytes 1-4
 Macrophages/ Histiocyte or osteoclast
Neutrophils & Precursors 53-63
o with storage iron – stain w/ iron stain
Monocytes 0-2
(Prussian blue stain)
Eosinophi;s & Precursors 1-3
o with lipids (Gaucher cells)– monocyte or
Basophils & Precursors 0-1
macrophage w/ cytoplasm appearing a
Lymphocytes 8-12
crinkled tissue paper because of Plasma Cells 0-2
leucocerebrocytes)
 Mast Cells / Tissue Basophil
 Osteoblasts – immature bone cells, waterbug or MYELOID ERYTHROID RATIO
comet appearance - ratio between the granulocytic precursors &
 Osteoclasts/macrophages – for bone marrow erythroid precursors but only the nucleated
resorption precursors
- on smear, count all myeloid cells & erythroid
precursor which has 3 pronormoblast &
basophilic normoblast & 4 orthochromic
normoblast & for granulocyte only the
neutrophil, eosinophil & basophil
o M:E RATIO = 2 granulocyte:1 RBC – 4:1
o AVERAGE: 3:1

SMEAR PREPARATION FOR MALARIA


EXAMINATION: SPECIMEN
MARROW CELLULARITY
- EDTA-anticoagulated blood or fresh capillary
- Percentage of marrow space occupied by
blood collected before the initiation of
hematopoietic cells compared w/ fat (yellow or
treatment.
red marrow)
- At least two thick and two thin peripheral blood
 MARROW FAT: hematopoietic cell = 1 : 2 (adults)
films should be made.
 MYELOID: Erythroid (M:E) ratio = 2 : 1 - 4 : 1
 RED MARROW CELLULARITY a. THIN BLOOD SMEAR
o HYPERCELLULAR/HYPERPLASIA: - For morphologic examination and species
increased in one or more cell lines w/ identification and determination of Percent
few fats thus increase marrow arasitemia
production like in polycythemia vera - PREPARATION: same as wedge method
o A drop of blood w/ 2-3 mm & 1 cm away increase predominance of blast cells or
from the end & smear via wedge method immature cells
o First fixed in methanol then add distilled o The decrease of blast cells and increase
water to let RBC to hemolyze to of mature cells is called chronic
removed Hb then stained w/ pH of 7.2 leukemia
pH due to hemozoin pH & examine o Predominance of myeloblast is
o Cells are still intact & not overlapping associated with Acute Myeloid
thus observing morphology Leukemia
o Predominance of mature myelocyte is
b. THICK BLOOD SMEAR associated with Chronic Myeloid
- For screening purposes because cells are Leukemia
distorted - Blast cells are not easily identified because of
- PREPARATION: place 3 small drops or large drop their common characteristics (larger in size, large
of blood close together near one end of the slide. nucleus, smaller and deeply basophilic
Spread in a 25 centavo coin & dry. With one cytoplasm), cytochemical stains are used to
corner of a clean slide, stir the blood for about differentiate in the absence of CD marker
30 seconds to mix the three drops over an area - Acute Myeloid Leukemia can be further classified
approximately 1 to 2 cm in diameter knowing it is a stem cell
o First dehemoglobinized; air-dried then - French American British System classified
stained leukemia based on the morphology of the cells
after staining with Romanowski stain and their
c. STAINS FOR MALARIAL BLOOD SMEAR reaction to cytochemical stain
- Wright’s; Giemsa - FAB further classified ALL as ALL1, ALL2, ALL3.
While AML is classified into seven categories
CYTOCHEMICAL STAINS
(AML1-AML7)
CYTOCHEMISTRY - In AML:
- Defined as the microscopic study and o AML1-AML3 predominance cell is
identification of chemical constituents within Myeloblast, promyeolocyte myelocyte
individual cells. It is useful in the identification of (granulocytic series)
malignant cell types on the basis of cytoplasmic o AML4 and AML5 uses alpha naphthyl
or nuclear chemistry; cellular constituents that Chloroacetate esterase
are present in abnormal form or amount; lack of o AML4 stain positive in both Specific and
cellular constituent; and cells exhibiting non- specific esterase because it
functional abnormalities contains both granulocytic and
monocytic cells
LEUKEMIA o AML5- is the acute monocytic leukemia
- refers to group of disorders o AML6 is called Erythroid Leukemia,
- In the category of cell line, leukemia can be leukemic cells are precursors of
differentiated to either Myeloid or Lymphoid erythrocyte. Positive in Periodic acid
Leukemia Schiff
- In the basis of Blast cell o AML7 leukemia of megakaryocytes
o Pre-dominance of lymphoblast is called called acute megakaryoblastic leukemia
Acute Lymphocytic Leukemia - One example for Chronic Lymphocytic Leukemia
o Pre-dominance of mature with few is the Hairy Cell Leukemia which affects B-cells.
lymphoblast is called Chronic Hairy cells contain ACP.
Lymphocytic Leukemia o ACP isoenzyme is present in all blood
o Acute leukemia refers to a type of cells and can be inhibited by tartaric
leukemia with poorer prognosis with an acid except the isoenzyme number 5
which is the one present in hairy cell
leukemia
- In Chronic Myeloid Leukemia the predominant
cells are the mature myeloid cells especially the
granulocytes. CML is also called Chronic
Granulocytic Cells. Granulocytes can range from
50,00-300,000
- Having a very high WBC count can also be
observed in Leukemoid Reaction which is due to
underlying condition
- Both leukemoid reaction and chronic myeloid
leukemia are characterized by a very high WBC
count. However, Leukemoid Reaction is
reversible that when the underlying condition is
resolved the WBC will return to normal count.
CLM is a permanent condition. To differentiate
Leukemoid reaction from CML, alkaline
phosphatase is used.

ENZYMATIC STAINS
Peroxidase/Myeloperoxi - The stain itself does not contain the peroxidase enzyme. The peroxidase enzyme
dase (MPO) is inside the cell, the composition of the stain is the substrate for peroxidase
enzyme
- Myeloperoxidase is a normal constituent of the primary granules of the
myelocytic cells which is observe as early as promyeolocyte
- Stain marker for primary granules & auer rods (fused primary granules)
- Stain positive in AML but negative in ALL
- Differentiates AML from ALL
- Stain marker for immature myeloid cells
- Stain positive in granulocyte but not in lymphocytes
- Positive Activity: reddish-brown deposits (in cytoplasm of granulocytes and
monocytes)
- Note: Myeloperoxidase enzyme deteriorates; Stain should be done only on fresh
specimens
Alpha-Naphthyl Acetate - Use to detect the granulocyte of monocytic origin
Esterase & Alpha- - Non specific because it can be seen in other cells
Naphthyl Butyrate - The stain does not contain esterase but rather substrate for the enzyme
Esterase (Non-Specific - If the cell contains the enzyme it will catalyze the hydrolysis of the butyrate or
Esterases/NSE) acetate in the stain
- Unfixed sample can be used so long in the dark for as long as 2 weeks
- Marker for cells of Monocytic origin
- Other cells (+) : Megakaryocytes, Platelets, Histiocytes, Plasmacytes, some T-
lymphocytes
- Positive Activity: red-brown/dark red
Leukocyte/Neutrophil - Stains NEUTROPHILS (the only leukocytes that contain this activity)
Alkaline Phosphatase - Neutrophils contain various amount of ALP
(LAP/NAP) - Principle: Differential count involving neutrophils
- Differentiates Leukemoid reaction (LR) from Chronic myeloid leukemia (CML)
- Reference Value: 30 –185 LAP score
- Increase LAP score is observed in the following:
o during the last trimester of pregnancy - Hodgkin disease
o Polycythemia vera - Multiple myeloma
o Aplastic anemia - Obstructive jaundice
- KAPLOW’s Scoring (Count 100 cells and grade them as follows)
o 0 = no staining
o 1+ = faint & diffuse staining
o 2+ = pale moderate amount of blue staining
o 3+ = strong blue precipitate
o 4+ = deep blue or brilliant
- To compute multiply the counted neutrophils by their grade
- Normal to High indicates Leukemoid reaction
- Below than normal value to zero indicates CML
Tartaric Acid Resistant - For the diagnosis of Hairy cell leukemia (HCL)
Acid Phosphatase (TRAP) - General Principle: ACP is Detected in almost all blood cells but when treated with
tartaric acid it is inhibited except isoenzyme number 5
- Labile if unpreserved
- Sample should first be fixed and be stored at -20 C and can be used atleast 2
weeks
- Other Tartrate Resistant cells: Sezary cells; Histiocyte; T-cell of acute lymphocytic
leukemia
- Activity is indicated by purple to dark red granules in cytoplasm
Cyanide-Resistant - For identification of Eosinophilic components
Peroxidase - Positive Activity: brown

Naphthol ASD - Marker for mature & immature Neutrophil and mast cells
Chloroacetate Esterase - Substrate is the chloroacetate
(Specific Esterase) - Use to identify immature or primitive granulocyte ALM1-ALM3
- Positive Activity: bright red granules in cytoplasm
- This enzyme is stable and may last for months
Terminal - Stains DNA polymerase immunoperoxidase
Deoxyribonucleotidyl - Marker for immature Lymphoid cell
Transferase (TdT) - Positive in almost 90% cases of ALL
- Also useful in the detection of the “lymphoblastic transformation” of chronic
myeloid leukemia (CML)
- Detects blastic transformation of CML
o CML can transform into an acute leukemia
o Acute leukemia more than or equal to 30% blast cells
o CML can transform either AML or ALL. If the blast cells presence stain
positive in MPO stain and SBB it is AML while if it stain positive in TdT it
means the it transforms into ALL
Acid Phosphatase - Present in all hematopoietic cells and found in lysosomes
- Activity is indicated by purple to red granules
- Unstable, cannot be stored

NON-ENZYMATIC STAINS
Prussian Blue stain - Stains siderotic granules (for the diagnosis of sideroblastic anemia)
- Used to detect hemosiderin in urine
- Use to identify iron in the ferric state (Fe+3)
Periodic Acid Schiff (PAS) - Stain reacts w/ aldehyde groups in glycogen, mucoprotein, glycoproteins, and
high molecular weight carbohydrates
- Does not detect stains instead it combines with the aldehyde group of glycogen
- Positive to all blood cells except normal erythroblast or pronormoblast
- Positive erythroblast indicates that it is affected by AML6
- Positive in erythroblasts in Di Guglielmo disease / Erythroleukemia
- L1 and L2 also stain positive in PAS
- The positivity of L1 to Pas is described to be chunky or block-like positivity
- L2 is not that large as L1
- Negative Activity: bright fuchsia pink (Pattern of staining varies with each cell
type)
- Also differentiates Acute Lymphoblastic leukemias
- Peroxidase of eosinophil is resistance to cyanide.
- Cyanide resistance peroxidase is used to identify peroxidase of eosinophils
Sudan Black B (SBB) - Marker for phospholipids and lipids
- Has the same staining reaction as myeloperoxidase
- Gives the same information as Peroxidase in the interpretation
- Positive in AML
- Advantage over myeloperoxidase is that it is more stable, because
myeloperoxidase detects enzyme which is somewhat labile and may disappear in
prolong storage. Sample stained in SBB can give reliable result for months
- Differentiates AML from ALL
- (+) Result: Dark purple-black granules
- Notes:
 Can be done on stored specimens.
 More sensitive than chloroacetate in the demonstration of mature &
immature neutrophils and mast cells
Toluidine Blue O - A metachromatic stain
- Binds with mucopolysaccharides in blood cells
- For the recognition of basophils and mast cells
- (+) Result: Reddish-Violet
HEMATOLOGY 2

GLOSSARY CAPILLARIES
HEMOSTASIS
- smallest blood vessels w/ the thinnest lining or
- Stoppage of bleeding walls thus easily ruptured which may be caused
- To maintain normal fluidity of blood in vivo. by increase in blood pressure/hypertension
There must be a balance in hemostasis. If not leading to discoloration of the skin depending on
needed, the clot should not be formed, but when the amount of blood that deposited on the skin
needed, the clot should be formed.
NOTE:
- Imbalance in hemostasis system will lead to
Discoloration of the skin depends on the volume of
excessive bleeding/hemorrhage or excessive
the blood.
clotting or abnormal thrombosis.
- Bleeding can be external or internal, severe or
mild which is indicated by red purple or blue COLOR TRANSITION
black which occur in the vessels particularly in
- bluish black if blood volume is greater or bluish
the capillaries thus blood deposit on the skin and
reddish/purplish to greenish to dark brown or
causes discoloration of the skin
greenish brown to yellow
PETECHIAE
NOTE:
- purplish red, pinpoint hemorrhagic spots in the  Changes in the color of skin is associated with
skin caused by loss of capillary ability to the degradation of Hb wherein Hb is degraded
withstand normal blood pressure and trauma into globin & heme wherein heme is further
- diameter of <3mm broken down into iron & protoporphyrin &
protoporphyrin is oxidized into biliverdin
PURPURA (green) & further converted into bilirubin
(yellow)
- produced by hemorrhage of blood into small
 Petechiae, purpura & ecchymosis are
areas of skin, mucous membranes, and other indication of primary hemostasis
tissues
- Red-purple-brownish yellow
- diameter of >3 mm but <1 cm POSSIBLE CAUSES OF RUPTURE OF CAPILLARIES:
- larger than petechiae or a combination of
1. Increase in blood pressure or hydrostatic
petechiae
pressure
- simultaneous rupture of capillaries
2. Pressure exerted outside
ECCHYMOSIS/ BRUISE
DISORDERS AFFECTING THE INTEGRITY OF CAPILLARIES:
- a form of purpura in which blood escapes into
1. Platelet abnormalities
large areas of skin or mucous membranes, but
2. Blood vessel wall abnormalities
not into deep tissue
- Black/blue – greenish brown or yellow NOTE:
- diameter of >3 cm  Even during normal situations, there are gaps
- due increase rupture of capillaries but not on the formed in between the endothelial lining
visceral organs but directly on the skin which is made up of connected endothelial
cells
NOTE:  Whenever gaps formed, platelet functions to
All of these are caused by the breaks of small vessels aggregate to plug the gaps in order to
particularly the capillaries. maintain continuity of blood vessel thus
limiting blood loss. Upon aggregation,
platelets are consumed leading to increase
appearance of petechiae or platelet may not
HEMATOLOGY 2

properly perform its function due to platelet causing deep vein thrombosis wherein both
abnormalities causes obstruction.
 Petechiae and Purpura may occur without
platelet & blood vessel wall abnormalities DEEP VEIN THROMBOSIS

- vaso-obstruction causing the amount of blood


EPITAXIS- nosebleed flow to be limited & the tissue will suffer from
ischemia due to decrease O2
HEMARUTHROSIS
EMBOLUS/EMBOLISM
- leakage of blood into a joint cavity
- bleeding in the joints resulting to clot formation - some parts of the clot detach & there is increase
causing obstruction in the movement causing in thrombus causing increase in pressure causing
damage to the affected joint leading to crippling the lumen to be smaller & the heart
which occur in severe hemocoagulation compensates causing increase blood output &
deficiencies such as hemophilia pressure leading to rupture & detachment of clot
& the detach clot may obstruct smaller vessel
HEMATEMESIS
THROMBOSIS- formation, presence of a clot in a blood
- vomiting of blood vessel
- content of esophagus or stomach which is acidic
(HCl)  2 TYPES:
- brown color blood due to reaction of blood to o PHYSIOLOGIC – if activation of
HCl coagulation system to form a thrombus
- pH of <7.35 is normal
o PATHOLOGIC – inadvertent formation of
HEMOPTYSIS blood clot
- expectoration of blood secondary to NOTE:
hemorrhage in the larynx, trachea, bronchi, or CAUSES OF PATHOLOGIC THROMBOSIS:
lungs  TTP
- coughs out blood  Cholesterol Plaque formation seen in DM
- red color w/ pH of 7.35 – 7.45 which thickens causing the vessel to be
sclerotic resulting to less capacity of the vessel
SPUTUM- product of the goblet cells of the lungs to dilate & constrict thus tissue reaction occur
HEMATOMA causing obstruction leading to slower
circulation & this increases platelet activation
- a swelling or tumor in the tissues or a body cavity & will form into thrombus
that contains clotted blood  Varicose veins / superficial thrombosis
- clotted blood with swelling  Deep veins thrombosis
 Arterial thrombosis
TRANSFIXATION- through & through of needle causing
hematoma
HEMATURIA
THROMBUS/CLOT
- presence of intact red cells in the urine or simply
- In vivo blood clot causing vascular occlusion and blood in urine
tissue ischemia - red with intact RBC thus presence of turbidity or
- composed of mostly platelets occurring in the smoky red
arteries causing arterial thrombosis or fibrin & a
small amount of platelet occurring in vein HEMOGLOBINURIA- presence of hb in the urine with
clear red color.
HEMATOLOGY 2

METHEMOGLOBINURIA – methemoglobin in urine or 2 STAGES OF HEMOSTASIS


oxidized Hb in urine due to storage at room temperature
a. Primary – consist of vasoconstriction & plug
giving a color of chocolate brown urine
formation
MENORRHAGIA – excessive menstrual bleeding  Components: blood vessels (endothelial lining),
platelets
MELENA
b. Secondary – consist of fibrin clot formation
- Stool containing dark red or black blood due to  Component: Coagulation factors
oxidized Hb. (Fibrinogen/CF1 converted by thrombin from
- Bleeding in the upper GIT such as in stomach or prothrombin wherein the conversion of
upper part of small intestine. prothrombin to thrombin is caused by CF10a, 5a,
Ca & PL to fibrin), Inhibitors (deactivates further
HEMATOCHEZIA
action) & Fibrinolytic proteins
- The passage of blood in feces.
PLASMIN/FIBRONOLYSIN – dissolves/lysis clot
- Red color bleeding in lower GIT such as in rectum
or colon. FIBRINOLYSIS – process of dissolution of clot

OTHER CAUSES OF RED COLOR IN STOOL PROCOAGULANTS- Promoters of coagulation

 Tomato, red beets, dragon fruit, diet  Components: clotting factor for fibrin,
 NOTE: phospholipids, platelets for plug formation
o the passage of blood in feces
ANTICOAGULANTS- Prevents excessive formation of
o red color bleeding in lower GIT such as in
platelet plug
rectum or colon
 Components: Natural inhibitors (ATIII, proteins-
HEMOSTASIS IS A COMPLEX MECHANISM THAT:
C & S), Fibrinolysis
1. Retains the blood within the vascular system
RESULT OF BOTH:
during periods of injury.
- vessels constrict/ vasoconstriction/vasospasm a. Thrombosis – increase procoagulant, decrease
thus limiting blood passage leading to limited anticoagulant
blood loss b. Hemorrhage – decrease procoagulant, increase
anticoagulant
2. Localizes the reaction involved to the site of
injury. CAPILLARY FRAGILITY TEST
- Platelet plug formation by first adhering of - This test measures the ability of small capillaries
platelet & will release their content & will to retain blood when subjected to increased
aggregate causing plug formation thus stoppage hydrostatic pressure and anoxia.
of bleeding initially. - It is a non-specific evaluation to measure
capillary weakness and deficiencies in platelet
3. Repairs and re-establishes blood flow through number and function meaning when there is
the injured vessels abnormal platelet number, there is abnormal
- Fibrin clot formation by converting fibrinogen CFT this is due to thrombocytopenia or when
into fibrin causing fibrin clot formation which will there is abnormal plt count but normal plt
stabilized the plug formation by forming a function thus abnormal CFT
meshwork. - Decreased capillary resistance causes the
- Promoted by thrombospondin & platelet derived capillaries to rupture which leads to bleeding
factor for smooth muscle & connected tissue and formation of petechiae.
muscle repair thus dissolving fibrin.
HEMATOLOGY 2

- Requires exposure to increase hydrostatic HEMOSTASIS


pressure & anoxia by applying a positive VASCULAR SPASM
pressure via BP apparatus or Tourniquet using
- very first reaction to injury
Rumpel-Leede Tourniquet Test or Negative
- function of blood vessels
pressure from the outside using suction cup
- Measures fragility or stability of capillaries as NOTE:
well as a non-specific evaluation of platelet  Vascular spasm & platelet plug formation are
number & function under the primary hemostasis while the clot
RESULT: petechiae formation is under the secondary hemostasis
 The primary hemostatic response towards
RUMPEL-LEEDE TOURNIQUET TEST injury is immediate but since the platelet plug
1. Examine the forearm, hand, and fingers to make is unstable the effect is short term
certain that no petechiae are present.  Secondary hemostatic response is more
complex but the response is quite delayed but
2. With a blood pressure cuff, apply 100 mmHg
once the clot is formed & is stabilized it will be
pressure to upper arm. To those who do not
efficient in effect which is long term
have a blood pressure cuff, use a tourniquet or
 In primary hemostasis, the platelets clump
rubber/cloth strip instead. Apply the tourniquet together & in secondary hemostasis, fibrin
not too tight, not too loose to employ just monomers are formed & connected together
enough pressure. or polymerized thus forming a meshwork
3. Maintain pressure for 5 minutes. around the platelet aggregate thus stabilizing
4. Release cuff and wait for 5 – 10 minutes before the platelet aggregate
making a final reading.  In the presence of meshwork, some of the
5. Examine the forearm, hands and fingers for blood cell in the circulation (RBC) are trapped
petechiae. in the fibrin meshwork & add up to the bulk &
the larger the size of the clot, the more
NOTE: efficient it is as a plug
Disregard any petechiae within ½ inch of the
blood pressure cuff (tourniquet) because this
may be due to pinching of the skin by the cuff. THREE HEMOSTATIC COMPONENTS

EXTRAVASCULAR - Play a part in hemostasis by


6. Count the number of petechiae and roughly
COMPONENTS providing back pressure on the
interpret as follows: injured vessel causing some to
1+ A few petechial on the anterior part of the flow outside & the tissues will
forearm absorbed & the blood causing
2+ Many petechial on the anterior part of the swelling which will exert a back
forearm pressure preventing the further
release or extravasation of the
3+ Multiple petechial over the whole arm and back
blood
of the hand
4+ Confluent petechial on the arm and back of the
DEPENDS ON:
hand
a) BULK or amount of surrounding
tissue.
b) TYPE of tissue (skeletal muscles
will be absorbed more as
compared to lose connective
tissues).
c) TONE of the surrounding tissue
(tissues that lack tonicity will not
HEMATOLOGY 2

absorbed that much such as PRIMARY HEMOSTASIS SECONDARY


tissues of elderly) HEMOSTASIS
VASCULAR - Involves the blood vessels INVOLVES: INVOLVES: activation of a
COMPONENTS Vasoconstriction, Platelet series of plasma proteins
DEPENDS ON: plug, Platelet adhesion, in the coagulation system
a) SIZE of the blood vessels release reaction & until fibrin clot formation
b) AMOUNT of smooth muscle aggregation (final product)
within their wall ACTIVATED BY: ACTIVATED BY: large
c) INTEGRITY of the endothelial desquamation and small injuries to blood vessels
cell lining injuries to blood vessels and surrounding tissues
Procoagulant substances Tissue factor exposed on
ENDOTHELIAL LINING are exposed or released cell membranes or
- composed of endothelial cells by damage or activated circulation
INTRAVASCULAR - Includes cells, plasma proteins & endothelial cells in order
COMPONENTS fibrinolytic agents such as the to activate the secondary Ex.:
plasminogen, plasmin & tissues hemostasis 1. Collagen exposed to
plasminogen activator and the surrounding tissue,
inhibitor causes platelets to
Platelets and biochemical in the aggregate & adhere &
plasma. activate CF 12. COLLAGEN
is the chief component of
 Clots are actually the the connective tissues.
plasma not the RBC 2. Tissue Factor
 There are inhibitors for introduced to the
coagulation and circulation because of
fibrinolysis large injury to the tissues,
 PECAM & ICAM contains it will activate factor 7
adhesion molecules causing the in vivo
 Platelets are also coagulation
involved in 2nd COMPONENTS: vascular COMPONENTS: platelet
hemostasis by exposing intima & platelets & coagulation system
their surface for the Rapid, short-lived Delayed, long term
clotting factors to be response response
activated & react Ends with platelet plug Naturally occurring
formation inhibitors in blood will
Primary Hemostatic block activated
ARTERIES VEINS CAPILLARY Disorders: Blood vessel & coagulation factors so
SIZE Large Smallest; platelet disorder that widespread
easily coagulation does not
rupture occur.
LININGS 3 layers of 3 layers of Endothelium FIBRINOLYSIS – slow breakdown & removal of fibrin
endothelial endothelial (inner) and clot as healing of the injured vessel occurs
lining, lining, Epithelial
smooth smooth cells
muscle and muscle and SPECIAL FUNCTIONS OF VASCULAR ENDOTHELIUM
connective connective
1. MECHANICAL ABILITY
tissues tissues
WALL Thicker Thinnest a. Vasodilation
o Prostacyclin – induces
vasodilation ; produce by
endothelial cell
HEMATOLOGY 2

b. Vasoconstriction  PROCOAGULANT: during vascular damage


- 2 Substances that Promotes o Serotonin & thromboxane A2 –
Vasoconstriction: vasoconstriction
o Serotonin/5 hydroxytriptamine o Von Willebrand Factor – from alpha
– from dense granules granules which is involve in platelet
o Thromboxane A2 adhesion & in the endothelial cell they
are stored in the weibel palate
NOTE:
o ADAMTS-13 – cleaves ultra large von
Both serotonin & thromboxane A2 activates the
Willebrand factor which may adhere
surrounding tissues for it to constrict causing now
limited amount of blood to enter even in the absence of damage which
may lead to TTP
o P-selectin – produce by other WBC &
2. SYNTHESIS produce by endothelial cell ; enhances
- Aids in anticoagulation & procoagulation adhesion to damage
o Collagen & Tissue Factor exposed –
 ANTICOAGULANT: during intact skin ; by smooth enters the circulation during damage &
surface allowing blood cells to circulate be exposed wherein collagen will cause
smoothly platelets to adhere & aggregate &
o Prostacyclin & nitrous oxide – secreted activate coagulation factors such as
by intact endothelial cell; induces Factor 7 & 7a causing clotting factors to
vasodilation & inhibit platelet occur for tissue factor
aggregation o Fibrinolysis – produces tissue
o Heparan sulfate – Increases the activity plasminogen activator which converts
of antithrombin 3 (major inhibitor of plasminogen into plasmin however
thrombin excessive TPA causes excessive plasmin
o Tissue Factor Pathway Inhibitor – major causing excessive fibrinolysis thus
inhibitor of tissue factor/extrinsic PLASMINOGEN ACTIVATOR INHIBITOR is
pathway which is initiated by factor 12 activated to inhibit activation of
o Anti-thrombin 3 – from the liver; major plasminogen in order to decrease the
inhibitor of the entire coagulation synthesis of plasmin & THROMBIN
system ACTIVATABLE FIBRINOLYSIS INHIBITOR
o Thrombomodulin – Inhibitor of Factor inactivates fibrinolysis
2a; binds w/ thrombin thus inhibiting
thrombin & cause deactivation of
protein C which is the cofactor of protein
S. Protein C comes from the liver
produced as an inactive protein which is
inactivated by thrombin &
thrombomodulin. Protein C becomes an
inhibitor. Protein C, thrombomodulin &
thrombin combines to form a
trimolecular complex & with endothelial
cell as a receptor, Protein C becomes
activated
HEMATOLOGY 2

PRIMARY HEMOSTASIS: Components, Functions, ADHESION •P-selectin; Procoagulant


Disorders MOLECULES Intracellular
BLOOD VESSELS (Vascular Intima) Adhesion
Molecules
ANTITHROMBOTIC, FIBRINOLYTIC AND COAGULANT •Platelet
SUBSTANCES RELEASED FROM OR FOUND ON THE endothelial
SURFACE OF ENDOTHELIAL CELLS cell adhesion
molecules
SUBSTANCE ACTION HEMOSTATIC
(PECAMs)
ROLE
•Help cells
PROSTACYCLIN •Inhibits Anticoagulan stick to each
(PGI2) platelet t and to their
activation surroundings
•Stimulates
vasodilation
HEPARAN SULFATE Coats the Anticoagulan NOTE:
endothelial t  Vascular disorders affect hemostasis
cell surface  Bleeding occurs if the connection of the
and weakly endothelial lining is loose.
enhances  Connective tissues are disseminated in every
activity of part of the body. Thus, all tissues of the body
antithrombin with defective connective tissue will also be
-3 affected.
THROMBOMODULI •Endothelial Anticoagulan
N (ENDOTHELIAL surface t
PROTEIN C receptor for Fibrinolytic BLEEDING DISORDER CAUSED BY VASCULAR
RECEPTOR) thrombin DEFECTS
(binds & HEREDITARY CONNECTIVE TISSUE DEFECTS
inactivates
thrombin) 1. EHLERS-DANLOS SYNDROME
•Enhances - Sex-linked ; caused by a defect in the gene
anticoagulant responsible for peptidase enzyme production
and which converts procollagen to collagen (chief
fibrinolytic component of the connective tissues of blood
action of vessels)
protein C - Characterized by: Hyper-extensive joints and
TISSUE FACTOR Controls Anticoagulan hyperplastic skin
PATHWAY Activation of t - LABORATORY FINDINGS: Normal Coagulation
INHIBITOR the extrinsic Test & Platelet function studies
pathway
TISSUE Converts Fibrinolytic
2. MARFAN SYNDROME
PLASMINOGEN plasminogen
- Defect in fibrillin
ACTIVATOR to plasmin
ADENOSINE Stimulates Reduces - Characterized by: Elongated upper & lower
vasodilation blood flow digits.
rate
VON WILLEBRAND Required for Procoagulant 3. PSEUDOXANTHOMA ELASTICUM
FACTOR (VWF) platelet - Autosomal recessive ; defect in elastic tissue
WEIBEL-PALADE adhesion to - Characterized by: calcified & formed into
BODIES site of injury plaques thus affecting the ability of the vessel to
HEMATOLOGY 2

constrict & dilate and structurally abnormal 2. CONGENITAL HEMAGIOMATA (KASABACH-


Elastic fibers MERRITT SYNDROME)
- Yellowish plaque, observed on skin where the - Disorder associated with tumors composed of
skin is abnormally loose (neck) many large vessels ; benign & can be surgically
remove
ACQUIRED CONNECTIVE TISSUE DEFECT
- Formation of fibrin clots, platelets consumption
1. VITAMIN C DEFICIENCY (SCURVY/SCORBUTUS) and red cell destruction secondary to vascular
- Vitamin C deficiency common among children obstruction occur at the site of tumor in the
thus prone to petechial formation mucous membrane
- IMPORTANCE OF VITAMIN C: for the formation
ACQUIRED ALTERATIONS OF VESSEL WALL
of the intact structure of the vascular basement
STRUCTURE
membrane & in synthesis of collagen for 1. DIABETES MELLITUS
hydroxylation of Vitamin C’s proline & lysine - Deposition of glycosylated proteins leading to
- Characterized by: gingival bleeding; hemorrhage thickening of capillary basement membrane
into subcutaneous tissues and muscles; large plaques affecting often the capillaries of the
hemorrhagic areas may develop just below the renal glomeruli and retina
eyes; splinter-like hemorrhage may also appear
in the fingernail beds 2. AMYLOIDOSIS
- LABORATORY FINDINGS: CFT = usually positive - Excessive amyloid or cholesterol deposit in
small vessels thus forming plaques causing now
2. SENILE PURPURA inflammatory process leading to cellular
- Related with normal aging process; inflammation and activation of cellular
discoloration of the skin which may not inflammatory process leading to injury & the
disappear & will remain as age spot because normal vessel flow will contain thrombus
even the function of the macrophages which formation
supposed to remove it has also declined - Causes vasoobstruction thus less oxygenation &
- LABORATORY FINDINGS: CFT = positive ; may also lead to rupture resulting to stroke &
Bleeding time = normal or only slightly prolonged other condition
HEREDITARY ALTERATION OF VESSEL WALL ENDOTHELIAL DAMAGE
STRUCTURE
1. AUTOIMMUNE VASCULAR PURPURA
1. HEREDITARY HEMORRHAGIC TELAGIECTASIA
- Problem with antibodies
(RENDU-OSLER-WEBER SYNDROME)
- Common among Scandinavian population ;
A. DRUG INDUCED PURPURA
discoloration of the skin where the skin is thin ;
- Common drugs that induce purpura:
blood vessels are abnormally thin thus they
sulfonamides & iodides quinine, procaine,
dilate & will be seen as spots
penicillin, aspirin, sedatives, coumarin
- Autosomal dominant trait characterized by thin-
- Endothelial cell might no longer be recognized as
walled, focally disorganized and dilated blood
self-cells & the immune system will induce Ab
vessels with a discontinuous endothelium
against the endothelial cell & iodide or
appearing as red pinpoint lesions most
sulfonamide
commonly on the face, lips, tongue, mucous
- May be due to development of antibodies to
membranes of the mouth and nose, ears,
vessel walls components, development of
conjunctiva, palms and soles
immune complexes and change in vessel wall
- LABORATORY FINDINGS: bleeding time, platelet
permeability
function tests, capillary fragility test and
coagulation test are all normal
HEMATOLOGY 2

B. ALLERGIC PURPURA/ANAPHYLACTOID NOTE:


PURPURA  Only metamegakaryocyte has the ability to
- Associated with certain food and drugs, cold produce platelets
temperature, insect bites and vaccinations  The more nuclear lobes, the more platelets it
- HENOCHS PURPURA – associated with will produce
abdominal pain secondary to GIT hemorrhaging
- SCHONLEINS PURPURA – associated with joint,
4 FUNCTIONAL ZONES OF PLATELETS
especially in the knees, ankles and wrist

2. INFECTIOUS PURPURA a. PERIPHERAL ZONE


- Associated with bacterial ; viral, rickettsial & - Contains glycocalyx which contains
protozoal infections or substance it produces glycoproteins (4, 6, 9)
- Contains glycoprotein 1b/9 for platelet adhesion
PLATELETS & GP 2b/3a for platelet aggregation
CHARACTERISTICS:

- Cytoplasmic fragments from megakaryocytes b. ZOL GEL ZONE


- Contains gel components that maintains
Diameter 2-4 um & 1/8 the diameter of different organelles at their position & allows
RBC them to communicate with one another
Site of production Bone marrow - Contains the cytoskeleton of the cell which
2 Pools 1/3 in spleen maintains the size & shape of platelet for it to not
2/3 in circulation
collapse which is made up of microtubules and
Growth Factor Thrombopoietin
microfilaments, the actin & myosin forms the
Manner of Division Endomitosis – increase
contractile protein, thrombosthenin.
nuclear component while
cytoplasmic component is
absent c. ORGANELLE ZONE
Rule As it matures, the cell size - Contains the dense granules, alpha granules
increases (most abundant), mitochondria & lysosomes
Mean Platelet 8-10 fL
Volume (MPV) d. MEMBRANE SYSTEM
Reference Platelet 150,000-450,000/ uL o Open Canalicular System – Membrane
count that allows communication to the
Daily Turnover 35 X 109/L (+/- 4.3) outside environment & route of entry &
Average production 2,000 – 4,000/megakaryocyte exit of substances
Lifespan 8-11 days o Dense Tubular System – for calcium
Function Maintenance of vascular pump & arachidonic acid production
integrity and blood
coagulation

DEMARCATING MEMBRANE SYSTEM

- Demarcation lines in cytoplasm ; where platelet


fragmentation occurs
- Appears as early as megakaryoblast & prominent
in promegakaryocyte & absent in megakaryocyte
& metamegakaryocyte.
HEMATOLOGY 2

PLATELET FACTORS

PF1 Plasma coagulation factor V


PF2 A globulin that inhibits antithrombin III;
increases platelet aggregation and
accelerates interaction of thrombin and
fibrinogen (fibrinoplastic platelet factor)
PF3 A lipoprotein (platelet phospholipid) found
in platelet granules & membrane and
required in 2 steps of the coagulation
process ; functions for coagulation factors to
assemble in order to cause fibrin formation
; component of prothrombinase together
w/ CF10, 5a, Ca for conversion of
prothrombin into thrombin for secondary
hemostasis ; assembly molecule
PF4 A glycoprotein stored in the alpha granules
and is extruded during the platelet release
reaction; aids in ADP-induced platelet
aggregation and inhibits effect of heparin

NOTE:
 In heparin therapy, patients might
develop autoantibodies against PF4
in Heparin Induce
Thrombocytopenia disease
 Heparin Induce Thrombocytopenia
– Acquired condition caused by
autoimmune disorder caused by
heparin therapy
PF5 Platelet fibrinogen
PF6 A plasma inhibitor associated with platelets
PF7 Cothromboplastin
PF8 Antithromboplastin factor
PF9 Accelator globulin stabilizing factor
PF10 Serotonin found in the dense granules
HEMATOLOGY 2

ROLE IN SUBSTANCE SOURCE PRINCIPAL FUNCTION/COMMENTS


HOMEOSTASIS
Promote HMWK (high molecular Alpha Granules  Contact Activation of intrinsic coagulation
coagulation weight kininogen) pathway
 Cofactor of 12a in converting prokalycrine into
kalycrine
Fibrinogen  Converted to fibrin for clot formation
Factor V  Cofactor to Factor 10 to convert prothrombin
into thrombin in fibrin clot formation
Von Willebrand Factor  Assists platelet adhesion to subendothelium
to provide coagulation surface
Promote ADP (strong) Dense bodies
Aggregation Calcium  Promote platelet aggregation
Platelet factor 4 Alpha granules  Calcium is involved in coagulation
Thrombospondin  Thrombospondin & platelet factor 4 is for
platelet function
Promote Serotonin Dense bodies
Vasoconstriction Thromboaxane A2 Membrane  Promotes vasoconstriction at injury site
precursors (inhibited phospholipids  Thromboxane A2 is a very strong aggregator
by aspirin therapy) of platelet
Promote Platelet derived Alpha granules  Promotes smooth muscle growth
vascular repair growth factor  For vessel repair
Beta-thromboglobulin  Chemotactic for fibroblasts
 For vessel repair
Other systems Plasminogen (from Alpha granules  Precursor to plasmin, which induces clot lysis
affected liver converted into
plasmin by TPA &
inhibited by A2-
antiplasmin)
A2 – antiplasmin  Plasmin inhibitor, inhibits clot lysis
C1 esterase inhibitor  Complement system inhibitor

PHYSICAL PROPERTIES AND FUNCTION OF - Von Willebrand Factor is necessary for areas
PLATELETS where circulation is with high shear (arteries &
1. ADHESION (platelet-to-injury) arterioles) such that if the platelets will simply
- Platelets during injury first cling & roll on the adhere, It will be easily dislodge but in veins &
damage endothelium & adhere on damage capillaries, since the pressure is not that strong,
endothelium the platelets can directly adhere on the exposed
- Sticking of platelets to a non-platelet structure; surface such as GP 6
platelets adhere only on detached or injured - Reversible.
endothelium
- Requires the GP Ib/IX in complex with V for 2. PLATELET RELEASE REACTION
adhesion which serves as a receptor for Von - Platelet undergoes shape or morphological
Willebrand Factor which is readily available in change. Alpha & dense granules release
plasma substances that will contribute to platelet
- Plasma Von Willebrand Factor is used for aggregation and activation of the coagulation
platelet attachment to the exposed system.
subendothelial matrix such as the collagen
HEMATOLOGY 2

- Releases ADP from dense for aggregation, 2. VON WILLEBRAND DISEASE


serotonin from dense for vasoconstriction & - Most common coagulopathy
thromboxane A2 from membrane phospholipid - Autosomal inheritance & function at
for vasoconstriction & aggregation chromosome 12 synthesized by the endothelial
- Irreversible cell & platelets; stable.
- Autosomal inheritance in Chromosome 12;
3. PLATELET AGGREGATION (platelet-to-platelet) deficiency in plasma VIIIc: VWF.
- Requires Gp IIb-IIIa as a receptor for fibrinogen - Characterized by prolonged bleeding time &
and plasma fibrinogen/Factor 1 which serves as same aggregation test result with Bernard-
glue or adhesive for platelet to platelet adhesion Soulier; prolonged APTT due to deficiency or
- Viscous metamorphosis decrease in CF 8c
- INITIAL AGGREGATION: reversible - FACTOR 8C/ANTIHEMOPHILIC FACTOR –
- SECONDARY AGGREGATION: irreversible cofactor in the intrinsic pathway
- AGGREGATING AGENTS: ADP, Thrombin, - ACTIVATED PARTIAL THROMBOPLASTIN TIME –
Collagen, Thromboxane A2, Epinephrine, test for intrinsic pathway
Arachidonic Acid, Ristocetin, Thrombospondin, - Multimeric molecule w/ a receptor for GP
Reptilase (from reptile) Ib/IX/V complex & collagen which is used to
bind w/ platelets causing platelet adhesion
OTHER FUCNTIONS:
- Contains Ag epitope & has a receptor for & a
4. VASOCONSTRICTION protein carrier for FACTOR
- Enhanced by serotonin released from platelets & 8C/ANTIHAEMOPHILIC 8C which is a sex-linked
thromboxane A2 from membrane phospholipids inheritance in the X chromosome synthesized by
- Reaction in primary hemostasis the liver & is labile thus it is prone to proteolysis
thus causing secondary deficiency causing
5. CLOT FORMATION prolongation of APTT but becomes relatively
- Platelet factor 3 or the assembly molecule is stable w/ Von Willebrand Factor.
needed for the formation of active plasma
ANTIHEMOPHILIC FACTORS:
thromboplastin
- Reaction in secondary reaction  A – Factor VIII (2 components: 8:VWF for
adhesion & 8C for coagulation intrinsic pathway)
QUALITATIVE PLATELETS (Platelet-vessel wall
 B – Factor IX
interaction)
A. ADHESION DEFECTS:  C – Factor XI

LABORATORY: PLATELET COUNT AND MORPHOLOGY:


1. BERNARD-SOULIER SYNDROME generally NORMAL
- Features:
o Deficiency in Gp 1b/IX thus an intrinsic COMMON SIGN: mucocutaneous bleeding
defect BLEEDING TIME (severe) & APTT TEST (prolonged) –
o Platelets are abnormally low & abnormal screening test
morphology
o Abnormally large platelet/giant platelet DIAGNOSIS: STANDARD VMD TEST PANEL TESTS:
w/ thrombocytopenia & prolonged
1. Quantitative VWF test (VWF Ag assay) – to
bleeding time
determine if VWF is quantitative or qualitative.
o AGGREGATION STUDIES: platelet
2. VWF activity test/ VWF RCo assay – qualitative
 Abnormal: Ristocetin
test to determine ability of vWF to bind to
 Normal: ADP, collagen &
platelets.
epinephrine
HEMATOLOGY 2

3. Factor VIII activity assay – to determine the


extent of VWF disease which may lead to
NOTE:
secondary deficiency of Factor 8
ACQUIRED VWF DEFICIENCY:
TREATMENT: - hypothyroidism, autoimmune,
lymphoproliperative & myeloproliferative,
 Cryoprecipitate – contains the complete disorders; benign monoclonal gammopathies;
component of factor VIII & VWF wherein this Wilms tumor ; intestinal angiodysplasoa;
control bleeding by supplying the deficient congenital heart disease ; pesticide exposure;
Factor VIII & VWF but it is no longer & hemolytic uremic syndrome
recommended because it does not undergo viral
inactivation wherein the specimen source is the
B. AGGREGATION DEFECTS (Platelet-platelet
blood donor.
interaction)
 Desmopressin acetate (1-desamino-8-D
arginine vasopressin/ DDAVP) – standard
1. GLANSMANN’S THROMBASTHENIA
treatment which cause release of VWF & Factor
- Features:
8 from endogenous sources in order to prevent
o Deficiency/ Gp IIb-IIIa; Inherited as an
bleeding.
autosomal recessive trait
 A plasma-derived factor VIII/VWF concentrate
o Bleeding time is prolonged, platelets are
CLASSIFICATIONS OF VON WILLEBRAND DISEASE morphologically normal but isolated w/
one another
TYPES DESCRIPTION o AGGREGATION STUDIES: Platelets
1  Partial quantitative deficiency of von aggregate normally with Ristocetin but
Willebrand factor (vWF) not with Epinephrine, ADP, Collagen
 Most common to occur about 75 -80% ;
a quantitative disorder
2. (CONGENITAL) AFRIBRINOGENEMIA OR
2  Qualitative (Function) deficiency of vWF
HYPOFIBRINOGENEMIA
 Level of vWF is normal but function is
o AFIBRINOGENEMIA: Acquired/Inherited
abnormal
total absence or severe deficiency in
2A  Decreased platelet-dependent vWF
fibrinogen.
function with selective deficiency of high-
molecular-weight multimers (the vWF is o HYPOFIBRINOGENEMIA:
more susceptible to proteolysis by Acquired/Inherited slight decrease in
ADAMTS-13 which is secreted by fibrinogen.
endothelial cells which destroys low o DYSFIBRINOGENEMIA: Inherited lack of
molecular weight vWF) fibrinogenemia
2B  Increased affinity for platelet
glycoprotein lb/IX/V DISORDERS OF PLATELET SECRETION & SIGNAL
TRANSDUCTION DEFICIENCY OF GRANULES
2M  Decreased platelet receptor binding
(STORAGE POOL DISORDERS)
(glycoprotein lb/IX/V)
ALPHA GRANULES DEFICIENCY
2N  (Normandy Variant) Impaired factor VIII
binding site leading to deficiency to APTT 1. GRAY PLATELET SYNDROME
thus APTT is prolonged - Deficiency of alpha granules
 Autosomal Hemophilia (royal disease) - Platelets aggregate abnormally & are abnormally
3  VWF is absent or nearly absent from large
plasma - Characterized by larger platelets colored gray to
 Rarest vWF & severe or complete blue-gray, and thrombocytopenia
deficiency
- LABORATORY: Prolonged Bleeding time;
 Autosomal Recessive
decreased aggregation with all agents.
HEMATOLOGY 2

2. QUEBEC PLATELET DISORDER DISORDERS OF PLATELET PROCOAGULANT


- Autosomal dominant disorder characterized by ACTIVITIES
abnormal proteolysis (destruction of proteins) & - In resting platelets, phosphatidylserine (PS) &
deficiency of alpha granules Phosphatidylethanolamine (PE) are located
- Abnormal increase in urokinase-type predominantly on the inner leaflet while
Plasminogen Activator phosphatidylcholine has the opposite
- Platelet count is normal or decrease distribution & this arrangement is maintained
- Characterized by delayed bleeding when by Aminophospholipid Translocase Enzyme.
exposed to trauma (12-24 hrs from the time of During platelet activation, it undergoes a
exposure) from mucocutaneous bleeding. morphological change such that its cytoplasm
will be extruded with extensions & the
DENSE GRANULES DEFICIENCIES distribution of phospholipids will be scrambled
1. WISKOTT-ALDRICH SYNDROME wherein phosphatidyl serine &
- (Sex-linked) deficiency of dense granules, with a phosphatidylethanolamine will be exposed &
defect in cytoskeletal assembly. acted by the enzyme, phosphatidyl
- Characterized by predominance of small scramblase/PF 3 & coagulation factors will be
platelets activated & be assembled
- TRIAD OF SYMPTOMS: thrombocytopenia,
recurrent infection & eczema 1. SCOTT SYNDROME
- Surface expression of phosphatidylserine is
2. HERMANSKY-PUDLAK SYNDROME decreased
(AUTOSOMAL) - Disorder of calcium-induced membrane
- TRIAD OF SYMPTOM: oculocutaneous albinism, phospholipid scrambling that when a platelet is
accumulation of ceroid-like pigment in activated it can assume a resting phase of
macrophages & bleeding tendencies platelet & may also be activated wherein the
exposure of phosphatidyl serine is deficient thus
3. CHEDIAK-HIGASHI (AUTOSOMAL) scrambling occurs & affects the secondary
- TRIAD OF SYMPTOM: albinism (no pigment), hemostasis
recurrent infection (immune deficiency) & giant
lysosomes 2. STORMORKEN SYNDROME
- Platelets are always in an activated state without
4. THROMBOCYTOPENIA WITH ABSENT RADII prior activation
SYNDROME (TAR) - Defect in aminophospholipid translocase
- Reduced radial bone or totally absent ; - Phosphatidyl choline is always on the outside
appearing like a t-rex syndrome causing abnormal platelet activation
- Structural defect in beta granules with
NOTE:
corresponding abnormal aggregation responses
 Phosphatidyl Choline – major component of
and thrombocytopenia the outer layer
- Characterized by severe neonatal  Phosphatidyl serine &
thrombocytopenia and congenital absence or Phosphatidylethanolamine – internal later
extreme hypoplasia of the radial bones (most
pronounced skeletal abnormality), cardiac and
other skeletal abnormalities
- Abnormal aggregation
- Observed as early as birth
HEMATOLOGY 2

ACQUIRED DEFECTS OF PLATELET FUNCTION o >450, 000: thrombocytosis


1. DRUG-RELATED  < 100,000/ uL – ABNORMALLY LOW; significant
 ASPIRIN – permanent cyclooxygenase inhibition decrease ; bleeding is not apparent
which is needed in the synthesis of thromboxane  30,000 – 50,000/ uL – BLEEDING IS POSSIBLE
A2 thus affecting platelet aggregation especially when exposed to trauma or bleeding
- Phospholipid A2 will convert the membrane secondary to surgical procedures
phospholipid of platelets into cyclooxygenase  <30,000 – SPONTANEOUS BLEEDING, meaning
- Prevents conversion of arachidonic acid into even without trauma or surgical procedure,
prostaglandin by cyclooxygenase leading to bleeding may occur in the internal organs
deficiency in thromboxane A2  <5,000/ uL – SEVERE SPONTANEOUS BLEEDING,
 NSAIDs and other COX-2 inhibitors (naproxen may cause intracranial bleeding and bleeding in
and ibuprofen) visceral organ due to inability of platelets to
 Other Antiplatelet agents: TICLOPIDINE AND maintain the continuity of blood vessels ;
CLOPIDROGEL – blood thinning drugs making considered as a life-threatening condition
blood viscous ; affects fibrinogen & Gp2b/3a
THROMBOCYTOPENIA
 Dextran – coating of platelets thus platelet
Causes:
cannot adhere & aggregate
1. DECREASED PRODUCTION
2. PARAPROTEINEMIAS ((MULTIPLE MYELOMA - Bone marrow defect, where platelet production
AND WALDENSTROM MACROGLOBULINEMIA) remains abnormal
- Dysproteneimias – Excessive plasma proteins to  MEGAKARYOCYTE HYPO-PROLIFERATION in:
lead to hyper-viscosity syndrome making the o APLASTIC ANEMIA – affects the bone
circulation slow leading to thrombosis & the marrow; bone marrow is infiltrated with
pressure is increase fats which are haematopoietically
- Disorder of the bone marrow thus production of inactive; all blood cells are decrease.
intrinsically abnormal platelets o MAY-HEGGLIN – inherited condition;
- Increase in bens jones protein & abnormally large megakaryocyte but are
immunoglobulins wherein for MM it is increase decrease in number; WBC w/ dohle
IgG while for WM it is increase IgM bodies.
o TOXIC CONDITIONS – when the toxic
3. RENAL DISEASE agents destroy megakaryocyte
- Uremia: increase in non-protein nitrogenous
substances ; leads to decreased thromboxane  INFECTIVE THROMBOPOIESIS – Infective
synthesis causing platelet destruction ; thrombopoiesis due to decrease in
Decrease: adhesion, platelet release, and thrombopoietin which is mainly produced by the
aggregation ; Increase: non-protein nitrogenous liver and a few in kidneys ; associated among
substances chronic alcoholics among ethanol abused thus
damaging platelet production
QUANTITATIVE PLATELET DISORDERS
- Platelets function is normal but platelets are  NEONATAL THROMBOCYTOPENIA – among
few or increased in number newborns who are exposed to infection inside
the uterus ; associated with infections with
SIGNIFICANT PLATELET LEVELS (platelet count and toxoplasma, rubella, cytomegalovirus, and
clinical manifestation): herpes and HIV (TORCH); and in-utero exposure
to certain drugs (particularly chlorothiazide
 150 – 450 x 109/L – REFERENCE PLATELET
diuretics and the oral hypoglycemic
COUNT
tolbutamide)
o <150, 000: thrombocytopenia
HEMATOLOGY 2

 MARROW REPLACEMENT/MYELOPTHISIS – 2. DISSEMINATED INTRAVASCULAR


replacement of entire bone marrow with COAGULATION (DIC) – patient produces a
abnormal cells such as cancerous cells substance that will activate both coagulation
and fibrinolysis leading to imbalance in
2. BLOOD TRANSFUSION coagulation as clotting occurs there is a
- Transfused platelets will add on the patients corresponding fibrinolysis, when clot occurs
platelets improving now the patients platelet some coagulation factors are consumed
count such as the CF 1, 5,8 and 13 and platelets ;
- Occurs after transfusion of platelet-containing occurs elsewhere the body ; involves the
blood products (recipient’s plasma is found to activation of both coagulation and
contain alloantibodies to antigens on the fibrinolysis due to liberation of
platelets of the transfused blood product) thromboplastic substance by damaged or
- Normal lifespan of platelets in blood banks is abnormal cells
only 5 days
 DILUTIONAL LOSS – blood product transfused 3. HEMOLYTIC URENIC SYNDROME (HUS) –
does not contain enough viable platelets thus it’s associated with diarrheal or non-diarrheal
effect is to dilute only the patients’ blood and a. DIARRHEAL among children after
improve the blood volume ; Massive transfusion children has been infected with GIT
where patient receives 4 or more units of blood problem with any organism that can
for 24 hours may also lead to dilution of patient’s produce a shiga-like organism the
platelets ; transfusion of intravenous fluids and verotoxin (E. coli H7:0157, Strep.
pure plasma Pneumoniae, shigella) where it will it
 POST TRANSFUSION PURPURA – occurs a week bind to glomerular capillaries on the
after ; related to antibody if the patient is already endothelial cells causing them to be
been sensitized or received blood component activated and become thrombotic
before causing now antibody production against promoting thrombosis; often occurs in
the platelets ; severe type of thrombocytopenia the kidneys ; It is hemolytic when
platelets become thrombus they cause
3. INCREASES DESTRUCTION OR CONSUMPTION lysis ; urine appears as pinkish ;
a. NON-IMMUNE Associated with mild febrile illnesses,
- premature activation of platelets in secondary certain immunizations, and
aggregation causing platelets to be consumed gastrointestinal disturbances (E.coli
- associated with activation and consumption O157:H7 or other Shiga/Vero toxin-
producing bacteria)
1. THROMBOTIC THROMBOCYTOPENIC b. NON-DIARRHEAL among children
PURPURA (TTP): caused by a deficiency of a associated with vaccination
ADAMTS-13 ; common in young adults ; 4. PLATELET LOSS ON ARTIFICIAL SURFACE – in
persistence of ultra large VWF which may cases of artificial heart valve, though it is
bind prematurely to platelets causing match, when platelets are in contact to
platelets to aggregate which is irreversible & them, platelets are destroyed
the clot may detach & may cause 5. INFECTIONS (eg. Dengue) – leads to platelet
vasooclussion; platelets are consumed when consumption, because dengue infects some
formed into thrombus ; deficiency in of the macrophages and monocytes which
disintegrin and metalloproteinase with a produces cytokines & the effect make the
thrombospondin type 1 motif, member 13 endothelial lining hyper permeable and
(ADAMTS-13) platelets will plug the area until platelet
count is consumed
HEMATOLOGY 2

b. IMMUNE DESTRUCTION 2. SECONDARY/ REACTIVE THROMBOCYTOSIS


- Associated with antibody (IgG) - due to underlying condition ; Seen in splenic
1. IDIOPATHIC/IMMUNE mobilization and in hemolytic anemias
THROMBOCYTOPENIC PURPURA (TTP) – a. SPLENIC MOBILIZATION – occurs in
cause is unknown for ITP; common among hemorrhagic conditions ; due to acute
young children ; due to presence of anti- hemorrhage, platelets moves into systemic
platelet antibodies of the IgG type circulation causing transient increase in the
associated with previous infection; most platelet circulation ; response to stress such
common in young females <15 y/o ; if occurs as in acute blood loss anemias ; platelets
in children, the prognosis improves but in formed and mature from the spleen
adults it is life threatening b. RAPID REGENERATION – involves the bone
2. DRUG-INDUCED: HEPARIN-INDUCED marrow but production is normal ; during
THROMBOCYTOPENIA (HIT)- patient hemolytic anemia where there is a need to
develops IgG antibody specific for heparin- loss of blood cells thus the bone marrow will
platelet factor 4 complexes; A. common side immediately try to compensate and increase
effect of unfractionated heparin production of blood cells ; platelets newly
administration released from the bone marrow indicated by
reticulated platelets in circulation in order to
4. SPLENIC SEQUESTRATION respond to underlying condition
- Seen in hypersplenism and splenomegaly where
LABORATORY EVALUATION OF PRIMARY
there is enlargement of spleen thus sequestering
HEMEOSTASIS
>1/3 of platelets & keeping 50-90% of platelets,
- Evaluates platelet and blood vessels.
leaving 10% of platelets in the circulation thus
leading to thrombocytopenia.
A. DIRECT METHOD OF PLATELET COUNT
- Can be performed directly or indirectly and can
5. UREMIA
also be done manually or with the use of
- Increase of NPN in the blood due to inability of
automated machines.
the kidney to remove these substances due to
- Manual Procedures for platelet count can be
renal insufficiency; also causes platelet damage.
done with the use of Light Microscopy Principle
THROMBOCYTOSIS/ THROMBOCYTHEMIA or the Phase-Contrast Microscopy Principle.
- Increases platelet count (>450,000/uL) - EDTA: Anticoagulant of choice whether
automated or manual procedure. It prevents
1. PRIMARY THROMBOCYTOSIS platelets from adhering and as well as
- main defect ; abnormal production ; seen in aggregating together.
myeloproliferative disorders where there is - When aggregation and adhesion occurs, this will
abnormal proliferation of blood result to a false decrease in platelet count.
a. POLYCYTHEMIA VERA – chronic However, even if the blood is EDTA
myeloproliferative disorder ; all blood cells anticoagulated, platelet count should be
are increase performed as early as possible. Preferable within
b. ESSENTIAL THROMBOCYTOSIS – increase the first 3-5 hours from collection.
platelets >1, 000, 000/uL w/ abnormal o 5 Hours: if blood is left in a room
function temperature (20°C). Otherwise, on
c. CHRONIC GRANULOCYTIC LEUKEMIA – prolonged standing, platelets will start
leads to abnormal marrow cell production to swell and break into smaller
fragment resulting to a false increase in
platelet count.
HEMATOLOGY 2

o 24 hours at 4°C: cell count (Platelet, RBC to provide moisture and prevent evaporation for
count, WBC count) are still valid if 15 minutes to allow the cells to settle
performed within the first 24 hours. d. After 15 minutes, place on the microscope &
start counting on the 5 small square or 25 central
AUTOMATED PRINCIPLE:
square or 10 small squares
- Counted within the volume range of 2-20 fL. All e. Compute:
cellular factors that falls within this value are
FORMULA:
considered by the machine as platelets. 𝑇𝑉−1
DF=
𝐵𝑙𝑜𝑜𝑑 𝑣𝑜𝑙𝑢𝑚𝑒
a. Electric impedance – platelets are counted in
1 𝑚𝑚3
triplicate (3x) VCF=
𝑛𝑜.𝑜𝑓 𝑠𝑞𝑢𝑎𝑟𝑒𝑠 ×𝑣𝑜𝑙𝑢𝑚𝑒 𝑜𝑓 𝑠𝑞𝑢𝑎𝑟𝑒𝑠
b. Light scattering

DILUENT: Plt. Count= no. of platelet counted × 𝑉𝐶𝐹 × DF

1. LIGHT MICROSCOPY METHOD (Tocantin


SOURCES OF ERRORS:
Method)
- Manual counting and requires additional stain 1. PLATELET CLUMPS – occurs if blood is not
- Principle: Hemocytometry immediately mix w/ anticoagulant during
- Counting chamber: improved Neubauer collection causing false decrease platelet count.
 Rees & Ecker Diluent: contains formalin as a 2. PLATELET SATELLITISM
fixative which preserves platelets and RBCs. It - Surrounding of platelets in a neutrophil.
also contains Sodium Citrate as an additional - Common among individuals who have develop
anticoagulant and a supravital stain which is the Ab (IgG) against EDTA.
Brillian Cresyl Blue (BCB). - Platelets attaches on WBC, band cells &
monocytes due to cell receptors on their cell
2. PHASE – CONTRAST MICROSCOPY METHOD surface.
- Identify cells based on their index of refraction. - Causes false decrease platelet count.
- Reference method for manual platelet counting. - Remedy: collect using Na citrate & perform
- Platelets are counted in 5R squares & appear as manual platelet count. However, platelet must
refractile or glistening bodies be multiplied by 1.1 to correct the dillutional
- Counting Chamber: Spencer-Bright line (S-B) effect of Na citrate to blood.
1475 3. EXCESS EDTA (1/2 full)
 Brecker Cronkite Method - Platelets will swell & fragmentize causing false
o 1% NH4 Oxalate diluent: no fixative and increase in platelet count.
stains. It removes RBCs from the - Fill EDTA half full (2.5 mL)
background wherein RBCs are 4. 3 HOURS AT RT – platelet will swell &
hemolyzed and platelets will remain in disintegrate causing false decrease platelet
the field. count thus, refrigeration of the sample at 4°C for
24 hours.
PROCEDURE FOR ALL:
PHYSIOLOGIC VARIATION:
a. First dispense 2-3 drops of the mixture before
loading the counting chamber 1. Decreased at birth
b. Dispense the mixture on both side of the 2. Increased after the 1st week of birth until adult
counting chamber normal values.
c. Place the loaded counting chamber on the wet 3. Decreased during menstruation.
house counting chamber which contains a
moistened cotton/filter paper w/ distilled water
HEMATOLOGY 2

B. INDIRECT METHOD release cuff & wait for 5-10 minutes before
reading.
1. FONO’S METHOD - The forearm, hands, and fingers are then
- DILUENT: 14% of MgSO4 – prevents platelets examined for petechiae 5 inches away from
from adhering & aggregating. blood pressure cuff.
- If there is a need to re-perform, perform on the
PLATELET ESTIMATION
other arm. It can only be repeated on the same
- The average number of platelets observed in 10 arm after a week.
fields & divided by 10 & multiplied by 20,000 in - NORMAL: 0 to occasional petechiae.
a blood smear anticoagulated with EDTA thus - INTERPRETATION: +, ++, +++, ++++
appearing clear & separated at the neck region
counted via cross sectional (side by side) or b. HESS/SUCTION TEST (Negative pressure)
battlefield method (serpentine) - A suction cup (2cm diameter) is placed in close
- In capillary puncture platelet appears as clump. contact with the skin at midpoint of upper arm
Thus, resulting to a false decrease of platelet for 1 min (pressure of 200-250 torr). An area
count. within a circle of 1 cm diameter is observed for
- Platelet shows 8-20 platelets per oil immersion petechiae 5 minutes after removal of suction
field of 200 RBC & 2-3 platelet clumps only and cup.
reported as follows: - CLINICAL SIGNIFICANCE: Positive in
thrombocytopenia, hypofibrinogenemia, and in
0-49,000 Marked decreased vascular purpura.
50,000- 99,000 Moderate decreased
100,000-149,000 Slight decreased TESTS FOR PLATELET FUNCTION & VASCULAR
150,000-199,000 Low normal FUNCTION
200,000-400,000 Normal 1. BLEEDING TIME
401,000-599,000 Slight increase - Measures the ability of the small blood vessels
600,000-800,000 Moderate increase to control bleeding after injury.
Above 800,000 Marked increased - Time it takes for a standard wound to stop
bleeding at a standard pressure (40mmHg)
- Standardized both incision & pressure applied
PLATELET & VASCULAR FUNCTION
- FACTORS AFFECTING BT:
1. TOURNIQUET TEST/CAPILLARY FRAGILITY TEST
a. Number of platelets and the ability of
- Tests the ability of small capillaries to retain
platelets to form plugs.
blood when subjected to increased hydrostatic
b. Thickness and vascularity of the skin.
pressure and anoxia.
c. Ability of the blood vessels to constrict.
- Affected by capillary integrity & platelet function
d. Severe coagulation factor deficiency causing
and number.
prolonged bleeding time.
- Becomes normal in presence of
- PROLONGED BT:
thrombocytopenia, hypofibrinogenemia &
o When platelet count is lower than 30-
vascular purpura
50,000/uL, or when platelets are
- Requires application of pressure which is applied
dysfunctional in VWD.
as a positive or negative pressure.
o After ingestion of aspirin/aspirin-
containing compounds, anti-
a. RUMPLE-LEEDE METHOD (positive pressure)
inflammatory, anticoagulants, and
- PRINCIPLE: an inflated blood pressure cuff on
some antibiotics.
the upper arm is used to apply pressure (100
mmHg) to the capillaries for 5 minutes. After,
HEMATOLOGY 2

2. PLATELET ADHESIVENESS/RETENTION TEST OTHER TESTS


- Reference Values: 26-60% CLOT RETRACTION TIME

- PRINCIPLE: When blood coagulation is complete,


a. GLASS BEAD METHOD/SALZMAN METHOD
clot normally undergoes retraction where clot
- Collect 2 whole blood samples, 1 using EDTA and
becomes denser and serum is expressed.
the other using glass bead collecting system.
Normally, clot retraction begins within 30
Perform platelet counts separately. (higher in
minutes after the blood has clotted &
EDTA)
completed within 24 hours.
- COMPUTATION:
- Normal clot retraction requires normal number
of functioning platelets. Ca, ATP, fibrinogen,
𝑝𝑙𝑡 𝑤𝑖𝑡ℎ𝑜𝑢𝑡 𝑔𝑙𝑎𝑠𝑠 𝑏𝑒𝑎𝑑𝑠−𝑝𝑙𝑡 𝑤𝑖𝑡ℎ 𝑔𝑙𝑎𝑠𝑠 𝑏𝑒𝑎𝑑𝑠 normal interaction of the platelets with
%= × 100
𝑝𝑙𝑎𝑡𝑒𝑙𝑒𝑡 𝑤𝑖𝑡ℎ𝑜𝑢𝑡 𝑔𝑙𝑎𝑠𝑠 𝑏𝑒𝑎𝑑𝑠
fibrinogen.
b. BORSHGERVINCT METHOD (in vivo test) - Thrombosthenin; ratio of the plasma volume
- Perform separate platelet count on venous and red cell mass; activity of a retraction-
blood and capillary blood samples promoting principle in serum & nature of the
- Formula: surface on which CRT is being measured.
𝑝𝑙𝑡 𝑐𝑜𝑢𝑛𝑡 𝑣𝑒𝑛𝑜𝑢𝑠 𝑏𝑙𝑜𝑜𝑑−𝑝𝑙𝑡 𝑐𝑜𝑢𝑛𝑡 𝑐𝑎𝑝𝑖𝑙𝑙𝑎𝑟𝑦 𝑏𝑙𝑜𝑜𝑑 CLINICAL SIGNIFICANCE:
%= × 100
𝑝𝑙𝑎𝑡𝑒𝑙𝑒𝑡 𝑐𝑜𝑢𝑛𝑡 𝑖𝑛 𝑣𝑒𝑛𝑜𝑢𝑠 𝑏𝑙𝑜𝑜𝑑
- CRT is poor when platelet count is less than
100,000/uL
3. PLATELET AGGREGATION TEST - in dysfibrinogenemia or hypofibrinogenemia,
- Aggregating agents; thrombin, arachidonic acid, paraprotinemias
ADP, collagen, epinephrine and ristocetin.
- Currently, this test is considered the gold a. HIRSCHBOECK METHOD (Castor oil method)
standard for evaluation of aspirin resistance. - Qualitative: test for the presence or absence of
- PRINCIPLE: Aggregating agents added to a retraction
stirred suspension of PRP induce a shape change - Formation of dimpling/droplet like serum on the
and aggregation of platelets. As a result, the PRP surface of blood drop.
changes from a turbid suspension to tone that - Normal values equals to 15-45 minutes.
transmits more light (clear) as the aggregates
are formed. The aggregometer records changes b. STEFANINI METHOD (test tube)
in optical density in the form of a graph. - Normal: clot retraction begins within 1 hour,
complete within 18 to 24 hours.
READING OF AGGREGATION RESPONSE:

1. PLATELET-RICH PLASMA AGGREGOMETRY: light c. MAC FARLANE METHOD


- Provides quantitative estimate of the degree of
transmittance aggregometer
2. WHOLE-BLOOD PLATELET AGGREGOMETRY: retraction.
- Normal values equals to 44 - 67%.
electrical impedance
𝑣𝑜𝑙𝑢𝑚𝑒 𝑜𝑓 𝑠𝑒𝑟𝑢𝑚
3. OPTICAL LUMI-AGGREGOMETER: for - %CRT= 𝑣𝑜𝑙𝑢𝑚𝑒 𝑜𝑓 × 100
𝑏𝑙𝑜𝑜𝑑
simultaneous measurement of platelet
aggregation and the secretion of ATP.
HEMATOLOGY 2

SECONDARY HEMOSTASIS: COMPONENTS, QUESTIONS TO REMEMBER:


PATHWAYS, TESTS; DISORDERS  Composition of Prothrombinase: Xa-Va-Ca++-
SECONDARY HEMOSTASIS PL
 Composition of the intrinsic tenase: IXa-VIIIa-
- involves coagulation factor deficiency Ca++-PL
(hemophilia A/B/C)  Factors present in fresh plasma: all are
- Depends on availability and functionality of present: no deterioration
different plasma coagulation factors.  Factors present in aged serum: II, VII, IX, X
(prothrombin); XI, XII, HMWK, PK (contact)
PLASMA FACTORS  Clotting → fresh serum: (-) I, II, V, VIII, XIII
- These are soluble plasma coagulation proteins  Storage: plasma: (-) V and VIII (labile factors)
that have crucial functions in the formation of a  Adsorption → absent in plasma(-): II, VII, IX,
X
clot. These proteins circulate as inactive
 Present in adsorbed plasma: I, V, VIII, XIII
zymogens (proenzymes) that become activated
(fibrinogen): XI, XII, HMWK, PK (contact
during the process of coagulation and, in turn,
group)
react with other factors in the system to  Most abundant: fibrinogen and prothrombin
ultimately generate thrombin which converts → less likely to be deficient.
fibrinogen to a localized fibrin clot.  Factor II → 80% convert to thrombin and only
20% remain in circulation
CLASSIFICATION OF COAGULATION FACTORS:
 Factors in the Common Pathway: I, II, V, X
 Substrate: are the substance upon which  Factors tested by both PT and APTT: I, V, II, X
enzymes act.  Fibrinogen Factors: I, V, VIII, XIII
 Coagulation factor deficiencies not related to
 Cofactors: accelerate the activities of the
bleeding: XII, PK, HMWK.
enzymes that are involved in the cascade. (TF,V,
VIII, HMWK)
 Enzymes: serine proteases and transaminase GENERAL RULE:
All coagulation factors are produced by the LIVER.
NOMENCLATURE:

- Except for Prekallikrein (PK) and HMWK which EXCEPT factors: VWF, III, and IV
are referred to by their name only, each factor
was assigned a Roman numeral number in order VON WILLEBRAND FACTOR (VWF)
of its discovery. The use of “a” that follows as
Roman numeral denotes the activated form. A - Produced by endothelial cells or endothelial
“f” refers to fragmented factor (e.g. XIIf) lining (Weibel palate bodies), platelets and
megakaryocytes (alpha granules).
GENERAL CHARACTERISTICS: (SUMMARY)
FACTOR III: THROMBOPLASTIN
 All are synthesized by the Liver except factors
VWF, III, and IV - Abundant: produced by all tissues in the body
 Deficiency in any factor may produce abnormal except blood cells.
hemostats; except of factors XII, PK and HMWK - When abnormally produced can trigger DIC
 Labile factors: V and VIII (short shelf life) patients release thromboplastin-like substance
 Factors prematurely activated by refrigerator that will activate both coagulation and
temperature. fibrinolysis.
- Crude extract from the tissue
- Component: tissue factor and phospholipid
- COMPLETE: Tissue factor and phospholipid
HEMATOLOGY 2

- PARTIAL/INCOMPLETE: phospholipid stabilization (cross-linkages) → fibrin as


component only insoluble clot
- PTT: reagent used is complete thromboplastin - Fibrin polymer: first clot formed (soluble and
- APTT: reagent used is phospholipid from partial unstable) unless factor XIII acted on it to stabilize
thromboplastin it.
o The one stabilizes the unstable and
FACTOR IV (CALCIUM)
soluble clot by forming covalent linkages
- Abundant in the plasma in ionized form. or cross-linking the fibrin monomers
- Can interact in coagulation mechanism. together.
- Sources: diet, bone, reabsorbed in the filtrate
NOTE:
ZYMOGENS: II, VII, IX, X, XI, XII, AND PK EXCEPT: Co-factors: V, VIII (need to be activated); III,
HMWK (need not to be activated)
 Zymogens – proenzymes: inactive enzymes; no Co-factors: increase activity of another coagulation
enzymatic function unless activated. factor
 II (Prothrombin): inactivated → IIa (thrombin) → Activated by thrombin: V → Va and VIII → VIIIa →
fibrinogen → fibrin; and activate other factors. function as cofactor.
 VII (stable factor: no fxn) → VIIa → activate Va cofactor of Xa → Prothrombinase (Xa-Va-Ca++- PL)
factor X and IX Xa → directly converts prothrombin to thrombin;
need Va to increase its function
 PK → kallikrein → enzymatic ability: serine
VIIIa cofactor of IXa → Intrinsic tenase (IXa-VIIIa-Ca++-
proteases
PL) needed to activate factor X to Xa
SERINE PROTEASES VIIIa: increases the activity of IXa in activating X
Factor III (Tissue factor) – cofactor of VIII- when
- When zymogen is activated and functions as combined to factor III, will be able to generate X and
serine proteases → which when activated, Xa
functions to activate the next enzyme or factor Extrinsic tenase: TF-VIIa → activate Factor X
in the pathway. HMWK: cofactor of XIIa; enhances XIIa activity
- e.g. XIIa → activate XI → XIa → IX → IXa → X → Function of XIIa: 1.) XIIa → PK → Kal; 2) activate XI
Xa → prothrombin → thrombin → XIa
NOTE:
NOTE: XII, PK, and HMWK deficiency – NO HEMOSTATIC
EXCEPT: I and XIII (BLEEDING) ABNORMALITY (IN VIVO STATEMENT)
- Factor I (fibrinogen): never activated (Note: never
write it as Ia) → write it as fibrin or fibrin monomer IN VITRO: Prolonged APTT
(activated)
e.g. thrombin once formed will activate fibrinogen to In some, PK and XII deficiency  thrombosis
become fibrin (wrong) → use the word cleaves (abnormal clot formation)
- Last to occur/be activated: cleavage of fibrinogen to
fibrin.
FIBRINOGEN: ultimate substrate in the coagulation NOTE:
mechanism (acted by thrombin). FACTOR I (FIBRINOGEN)
Cofactors: V, VIII, III, HMWK - First to be discovered and most abundant
- Fibrinogen deficiency is unlikely to occur.
- PLASMA PROTEIN (absent in serum due to
FACTOR XIII: FIBRIN STABILIZING FACTOR/LAKI- consumption → fibrin) → NOT a serum
LORAND FACTOR protein)
- (activated by thrombin) → transaminase or LABILE FACTORS: V and VIII (AHF-A)
transglutaminase enzyme → create covalent - Immediately disappear in stored plasma at
bonds between and among fibrin monomers → room or ref temperature (in vitro).
HEMATOLOGY 2

- Factor V (labile factor/proaccelerin) – most


labile.
- Factor VIII: labile in vitro and in vivo if it is not
being complexed with VWF.
- APTT (intrinsic pathway test): prolonged in
factor VIII deficiency and liver diseases.

FACTOR VII (STABLE FACTOR)


- IN VITRO: most stable → present in plasma
even if it is stored for several hours.
- IN VIVO: has shortest circulating life (3 hours)
and it is the first to disappear in the
circulation.
- In liver disease: first to be deficient
- FXN: in extrinsic pathway, it is the only
coagulation factor involved in extrinsic
pathway
- Prothrombin Time Test (extrinsic): prolonged
in VII deficiency and liver diseases.
- Prothrombin time: test that is more sensitive
in evaluating liver diseases, vitamin K
deficiency or antagonist.

PLASMA COAGULATION FACTORS (13)

- 12 coagulation factors assigned with Roman


numeral number (RNN) and named according to
order of their discovery.
- Two assigned with RNN but often referred by
their name: Factor III (Tissue factor or Tissue
thromboplastin) and IV (Ionized calcium).
- Factor VI: assigned to a discovered coagulation
factor (removed)  same as activated factor V
(Va): did not move/rearranged assignment of
coagulation number.

COAGULATION FACTOR NOT ASSIGNED WITH ROMAN


NUMERAL NUMBER:

1. HMWK (Kinin system)


2. PK (Kallikrein system)
- Belong and functions to other system.
- Also have a role in coagulation (minor).
3. Platelet factor 3: Platelet phospholipids →
exposed when platelets are activated:
phosphatidyl serine (major) and phosphatidyl
ethanolamine → assembly molecule →
formation of enzyme complex.
HEMATOLOGY 2

NUMERAL NO. PREFERRED NAME & FUNCTION BIOLOGIC HALF- MEAN


SYNONYM/S LIFE; REMARKS CONCENTRATION
I Fibrinogen Thrombin 2-4 days 200-400 mg/dl
substrate
II Prothrombin Serine protease 3-4 days 10 mg/dl
 Prethrombin
III Tissue Factor Cofactor Lipoprotein found in None
 Tissue Thromboplastin most of the body
tissues; insoluble
IV Calcium Mineral Ionized state 8-10 mg/dl
V Proaccelerin Cofactor 36 hrs 1 mg/dl
 Labile factor Most unstable &
 Accelerator globulin deteriorates
(AcG) Rapidly at RT
VII Proconvertin Serine protease 3-6 hrs; disappears 0.05 mg/dl
 Stable factor rapidly from the
 Serum Prothrombin blood when
Conversion Accelerator production is halted
 Autoprothrombin I (e.g. coumarin)
VIII:C Antihemophilic factor (AHF) Cofactor 10-14 hrs 0.01 mg/dl
 Antihemophilic globulin Bound to vWF
(AHG) (Factor VIII complex)
 Antihemophilic factor A
(AHF A)
 Platelet cofactor I
IX Plasma thromboplastin Serine protease 18-24 hrs 0.3 mg/dl
component (PTC)
 Christmas factor
 Antihemophilic factor B
(AHF B)
 Platelet cofactor 2
X Stuart-Prower factor Serine protease 40-60 hrs 1 mg/dl
 Stuart factor May be stored up to
 Prower factor 2 mons at 40C
 Autoprothrombin III
XI Plasma thromboplastin Serine protease 40-70 hrs 0.5 mg/dl
antecedent (PTA) Circulates in
 Antihemophilic factor X complex with
(AHF C) HMWK
XII Hageman factor Serine protease 50-70 hrs 3 mg/dl
 Glass factor Heat stable; can be
 Contact factor stored at 40C for 3
mons
XIII Fibrin stabilizing factor (FSF) Function? 11-14 days 2 mg/dl
 Laki-Lorand factor
 Fibrinase; Fibrinoligase
 Plasma
transglutaminase
Prekallikrein (PK) Serine Protease 35 hrs 35-50 mcg/ml
HEMATOLOGY 2

 Fletcher Factor
High Molecular Weight Kininogen (HMWK/HK) cofactor 9-10 hrs 5 mg/dl
 Fitzgerald factor; Contact activation
cofactor
 William factor; Flaujeac factor
Other Procoagulants:
VWF von Willebrand factor Factor VIII carrier; 24 hrs 1 mg/dl
Functions in platelet
adhesion
Platelet factor 3 Phospholipids- Assembly molecule Released by
primarily the platelets
Phosphatidylserine,
PF3

NOMENCLATURE FOR FACTOR VIII OF THE Test: Bleeding time test


INTERNATIONAL COMMITTEE ON THROMBOSIS AND Portion of molecule responsible for
HEMOSTASIS binding to endothelium and supporting
- In circulation, FACTOR VIII circulates in complex normal platelet adhesion and function.
with vWF. Tested by bleeding time
VIIIC: Ag Test: Immunologic monoclonal Ab
Antigenic property of procoagulant
2 MAJOR COMPOSITIONS portion as measured by immunologic
monoclonal antibody techniques
a. High Molecular Weight VWF (autosomal/
vWF: Ag Formerly known: VIIIR: Ag
chromosome 12)
Test: Laurell rocket or
- stable and serve as protein carrier for VIII and immunoradiometric factor assay
low molecular weight VIII:C (sex-linked in X Factor VIII-related antigen, which is a
chromosome) property of the large vWF portion of the
- labile if not carried by vWF  in plasma circulate molecules and measured by immunologic
in complex techniques of Laurell rocket or
b. Low Molecular Weight VIIIC (sex-linked in X immunoradiometric assay
chromosome) The antigen portion of the protein
- labile if not carried by vWF)  in plasma portion of vWF; now vWF:Ag
circulate in complex VIIIR:RCo Ristocetin cofactor activity
Allows platelets to interact or aggregate
VIII/vWF VIII:C and VWF with ristocetin
The molecule as it circulates in the plasma Ristocetin cofactor activity, which is
Composed of VIII:C and VIII:vWF portions factor VIII-related activity required for
non-covalently bound aggregation of human platelets with
VIII:C Function as a cofactor in intrinsic ristocetin in in vitro aggregation studies.
pathway coagulation (antibiotic no longer used
Test: Activated Partial Thromboplastin therapeutically)
Time (APTT/ PTT)
Portion of molecule acting in intrinsic
system as cofactor to IXa (with calcium) in NOTE:
the conversion of factor X to Xa. Tested by FACTOR DEFICIENCY- Leads to bleeding
partial thromboplastin time XII, PK & HMWK Deficiency- No bleeding abnormality
The pro-coagulant portion
VIII: vWF Function in primary hemostasis:
adhesion (platelet function)
HEMATOLOGY 2

THE 3 GROUPS OF COAGULATION FACTORS (12)


- Based on their common characteristics or
properties

1. FIBRINOGEN GROUP
- Factors: I, V, VIII, XIII
- They are large molecules
- Act as Substrate for Plasmin
- Consumed during the process of coagulation
- They are susceptible to denaturation (most
labile).
- Increase in concentration during an
inflammatory response.
- Increased in women using contraceptive pills
and in pregnancy

2. PROTHROMBIN GROUP (VIT.K DEPENDENT


GROUP)
- Factors II,VII,IX,X
- They are dependent on vitamin K for their
synthesis in the liver
- Oral anticoagulants: cause a functional decrease
in these factors by producing abnormal proteins
that cannot bind calcium
- They are not consumed during coagulation
- They are stable compounds that are well
preserved in stored plasma.
- They require calcium as a cofactor for binding to
phospholipids surfaces.
- Are readily adsorbed from the plasma by BaSO4,
or Al(OH)3.
- They are decreased in patients with problems in
gastrointestinal absorption

3. CONTACT GROUP
- Factors: XI,XII, Prekallikrein, HMWK
- They are fairly stable and not consumed during
coagulation.
- They require contact with negatively charged
surface for activation.
- They are closely related to fibrinolytic, kinin, and
complement systems.
HEMATOLOGY 2

Characteristics FIBRINOGEN GROUP PROTHROMBIN GROUP CONTACT GROUP


Members I, V, VIII, XIII II, VII, IX, X XI, XII, HMWK, PK
Consumed during CONSUMED NOT CONSUMED NOT CONSUMED
Coagulation/ Exception: Factor II-
clotting partially consumed

Factors in serum ABSENT in Serum PRESENT PRESENT


PRESENT in Plasma
Adsorbed by NOT ABSORBED ADSORBED NOT
BaSO4/ Al(OH)3

Factors present in PRESENT ABSENT PRESENT


BaSO4 adsorbed
plasma
Vit. K dependent NOT REQUIRED DEPENDENT NOT REQUIRED
LEVELS NOT AFFECTED
(For functional ability of the
factors)  binding on surface
of platelet or any phospholipid
Require Ca++ for NOT REQUIRES NOT
binding to PL Needs to be carboxylated in
surface order to interact with Ca++ in
the presence of vit. K
HIGHEST Molecular Weight
MOST LABILE STABLE STABLE
(V and VIII) Vitamin ADEK fat soluble
Absent in stored plasma: V vitamins  should be
and VIII properly digested bile for
Substrate to plasmin (main fat solubilisation PRESENT IN ALL SAMPLES
CHON for fibrinolysis) If there’s an obstructive
jaundice  bile can’t be
Thrombin cleaves factor I, V, drained in duodenum
VIII, XIII Undigested fats and vit K.
Found in platelets: will not be reabsorbed Closely related to fibrinolytic,
I and V: alpha granules kinin and complement systems
XIII – general cytoplasm of Decrease GIT absorption
the platelets (not stored in problems: (e.g. Obstructive 1. XII  XIIa = coagulation
granules) jaundice, excessive bacterial 2. XIIa  PK Kal  Kininogen
EXCEPT: VIII:C  plasma overgrowth) = Kinins
bound to VWF (protein 3. XIIa-Kal: plasminogen
carrier) Affected by oral activator  plasmin =
Increase in inflammation anticoagulants: immediate fibrinolysis
(APP) and pregnancy; in action: coumarin, 4. Plasmin  complement
women using contraceptive coumadin, warfarin - inhibit activation
(V and VIII) synthesis of vit. K; vit. K
antagonist
HEMATOLOGY 2

NOTE: THEORIES ON COAGULATION


ADSORPTION: process of removal of different
coagulation factor; adherence of smaller molecule to 1. MORAWITZ/CLASSICAL THEORY (1905)
larger molecule or surface. - Believes that there are 2 stages coagulation:
BaSO4/ Al(OH)3: large molecule unto which small a. Thrombin formation: by the combination of
molecules adhere prothrombin plus calcium ions plus tissue
Prothrombin Group: needs Vit. K for them to interact thrombokinase.
with Ca++ → bind with platelet PL
Excessive fibrinolysis: plasmin destroys intact factor V
and VIII

b. Fibrin formation: the formed thrombin acts


on the fibrinogen in the plasma.

2. HOWELL’S THEORY (1910)


- This theory believed that there are 5
coagulation factors involved: (Fibrinogen,
FACTORS II, VII, IX, X: Terminal AA (Glutamic acid): Calcium, Prothrombin, Anti-prothrombin,
requires additional –COOH (carboxyl radicals)  Thromboplastin)
gamma carboxylation (addition of another –COOH)
 to make it an anion (net negative charge)
interact with cation (Ca++) adhere or assembly on
the surface of platelet phospholipid (negatively
charge).
Gamma carboxylation: Vitamin K is required as a
cofactor for Factor IX to gain a negative charge for it
to attach with calcium.

Sequence of reactions:
NOTE:
1. Clotting (Serum): (-) I, V. VIII, XIII a. Prothrombin is inactivated by anti-
2. Adsorption: (-) II, VII, IX, X prothrombin/ heparin
3. Storage Age: (-) V and VIII b. When prothrombin needs to be activated
(vessel injury) → Thromboplastin neutralizes
anti-prothrombin and releases prothrombin
COAGUALATION MECHANISM c. Prothrombin → (Ca++ and thromboplastin) 
- Coagulation is a complex series of cascading Thrombin
reactions involving development of enzymes d. Thrombin + Fibrinogen → Fibrin
from their precursor. As one enzyme formed, it
converts the next zymogen to its activated 3. MODERN THEORIES: “WATERFALL” &
enzyme. This process continues until a fibrin “CASCADE” (1964)
meshwork clot has formed. Fibrin forms a - By Breckenridge and Ratnoff
meshwork in and around the primary hemostatic - 3 distinct phases:
platelet plug-like spider web, producing a stable
physical barrier.
HEMATOLOGY 2

 Activation of factor X via the intrinsic or extrinsic


pathway (start of COMMON PATHWAY).
 Conversion of prothrombin to thrombin
 Conversion of fibrinogen to fibrin
 Advocated the phases of coagulation

PHASES OF COAGULATION: Coagulation is divided


into four phases
1. Contact Activation Phase (XII – IX)
- Starts with the activation XII (comes in contact
with negative surface), then activation of
prekallikrein to kallikrein; and factor XI.
2. Activation of Factor X (via intrinsic Pathway or IN VITRO COAGULATION
via Extrinsic Pathway)
- Formation of Thrombin: Conversion of 1. INTRINSIC SYSTEM
Prothrombin to Thrombin - All necessary factors are IN the blood
3. Formation of Fibrin (circulation) VIII, IX, XI, XII, HMWK, PK
- All components are found in the circulating
COAGULATION CASCADE blood. To be activated, the blood must have
direct contact with a foreign object such as a
damaged blood vessel or glass. This results to
absorption of XII of the negatively-charged
collagen exposed by vessel wall damage.

Sequence of reactions:

a. Factor XII is activated by the foreign material


into XIIa;
b. XIIa reacts weakly with XI, prekallikrein (also by
XIIf), and plasminogen, thus converting them
into their forms
HEMATOLOGY 2

- Formed kallikrein feeds back to XII and causes  EXTRINSIC TENASE: TF + VIIa = TF/VIIa 
enzymatic cleavage producing more XIIa. activates X in common pathway
c. XIa in the presence of Ca activates IX  Tissue factor is necessary for the
activation of this system. All cells with
Note: IX can also be activated by VIIa-Ca-TF complex:
the possible exception of those in the
(Current concept of Coagualation)
blood, contain tissue factor. When
d. IXa in the presence of Ca, platelet phospholipids, injury occurs, the tissue factor is
and VIIIa activates X. released and acts as a cofactor in
NOTE: initiating coagulation. The released
APTT: deficiency in any factors  PROLONGED APTT tissue factor bind to VII and activates it
Activators: Exposed collagen (major component of to VIIa. Then, VIIa, together with
connective tissue that surrounds and support BV) calcium, activates X to Xa of the
and chemical  XII (contact factor)  XIIa  common pathway.
perform several functions:  VII  prothrombin time: deficiency of
XII (glass factor/contact factor)  XIIa
VII  prolonged PT
1. Coagulation cascade: 1. (CONTACT ACTIVATION
PHASE: Activate XI  IXa); (IXa-VIIIa –Ca++-PL:
Intrinsic tenase); convert PK to Kal 3. COMMON PATHWAY
2. Initiate fibrinolysis: XIIa  plasminogen to - Where both intrinsic and extrinsic pathway
plasmin usually meet or merge. The application is in in-
e.g. Which of the pathway occurs when the blood is vitro coagulation. Superseded by the current
placed on a tube or glass slide and allowed to clot? concept of the coagulation  REVISED
INTRINSIC PATHWAY  coagulation XII (glass factor) COAGULATION MECHANISM (In vitro:
 XIIa laboratory tests)

2. EXTRINSIC SYSTEM/ TISSUE FACTOR PATHWAY


- All factors needed are in the tissue NOT in the
circulation. Produce by all tissues but not by
blood cells. ONLY FACTOR VII is involved.
- Does not occur outside the body unless you add
a reagent
- Tissue Factor/Tissue thromboplastin
introduced in the circulation when there’s
vessel/tissue injury. - Begins with activation of factor X via the
- In classical concept: TF activates VII  VIIa and Intrinsic Pathway (IXa-VIIIa-Ca-PL) or via the
combines with VIIa Extrinsic Pathway (VIIa-III/TF) to form
- In current concept: VIIa (inert material) is Prothrombinase/Xa-Va-Ca-PL. This
already available in the circulation in small prothombinase complex converts prothrombin
amount (1-2%)  combine with TF/Factor III to thrombin which enzymatically cleaves
(cofactor) fibrinogen into fibrin.
- Thrombin activates XIII  XIIIa
(transaminase/transglutaminase)
- 4 FACTORS INVOLVED: X, V, II, I  tested by
PT/APTT.
HEMATOLOGY 2

NOTE:
In performing a laboratory test on coagulation, follow
and interpret the test using the CLASSICAL CONCEPT
OF COAGULATION

Routine Screening Test for coagulation factor


deficiency: COAGULATION TESTS: measure how long
does the plasma take to form a clot; end result 
clot formation
Intrinsic: APTT
Extrinsic: PTT (Prothrombin Time)
PT: prolonged; APTT: Normal  Extrinsic factor
 alpha, beta and gamma chains terminates in D-
deficiency (VII)
PT: Normal APTT: prolonged  Intrinsic factor domain and meet at the center coil E-domain
deficiency (XII, XI, IX, VIII, HMWK, PK)  exposing alpha and beta fibrinopeptides 
Both prolonged  Coagulation factor deficiency of thrombin (proteolytic enzyme) cleaves
the common pathway (X, V, II, I) fibrinopeptides A and B  removed  fibrin
In APTT/PTT (in vitro testing): XIII is not included  monomer (insoluble)  several formed 
not detected
polymerization: end to end and side to side
XIII not needed for the formation of clot  stabilize
the clot connection  fibrin polymer (clot) soluble and
unstable clot  fibrin cross-linking by factor
XIII  covalent linkages in D-terminal 
NOTE: stabilized fibrin
COAGULATION ROLES OF THROMBIN: activates V,
VIII, XIII, cleaves fibrinogen + activates XI. STEPS IN CONVERTING FIBRINOGEN TO FIBRIN
1. Thrombin cleaves the alpha and beta chains
only (releasing fibrinopeptides A and B) 
FIBRIN FORMATION
FIBRINOGEN STRUCTURE: soluble fibrin monomer/unstable gel.
2. Fibrin monomers aggregate spontaneously end
- D-terminal domains and a central-E domain to end, side to side to form soluble fibrin
- 3 types of polypeptide chain (pairs): polymers thus vulnerable to enzyme plasmin
o + Fibrinopeptides (2A) (also soluble in 5M urea).
o Alpha (2B) beta 3. Factor XIII: XIIIa + Ca crosslinks adjacent fibrin
o Gamma (no fibrinopeptide) monomers by forming covalent bonds to form
- Fibrinopeptide: removed by thrombin stabilized fibrin.
- Fibrinogen: soluble protein; not seen in plasma,
but once fibrinopeptide A and B is removed  OTHER RELATED SYSTEMS:
insoluble (clot is seen). 1. Kinin System: this system is involved in
chemotaxis, pain sensation, and mediate
inflammatory responses, increased vascular
permeability, and vasoconstriction and induce
smooth muscle contraction.
2. Fibrinolytic system: The Kallikrein, XIIa, &
HMWK act as plasminogen activator.
3. Complement System: this mediate immune and
allergic responses.
HEMATOLOGY 2

CLASSICAL (IN VITRO) FACTOR XII: In vivo coagulation; their role is not
necessary; can be by passed; XI can be activated by
- Does not explain how physiologic hemostasis thrombin; deficiency will not cause bleeding
occur in vivo. conditions.
- Interdependent pathways

IN VIVO CONCEPT OF COAGULATION/ CURRENT


CONCEPT/ REVISED CONCEPT OF COAGULATION
- Current Concept of the Coagulation Cascade (In
vivo coagulation)
Key initiating step: cause to start the reaction:
- INITIATING step: exposure of TF and its reaction
activation of XII and VII (more important to occur);
with VII. VIIa/TF  series of reaction
2 FACTORS ACTIVATED BY TF/VIIa: X and XI  Xa and IIa (positive feedback activity: once formed go
production of thrombin back to their initial reaction and proceed to cause
more activation)
a. INITIATION: Extrinsic tenase  3%- 5% lIa  Xa  VII  VIIa (2 chain) more effective in
- Small effect of activation of IX and X + Tissue function
Factor Pathway Inhibitor (naturally occurring  VIIa (single chain) produce by other factors or in
inhibitor) inhibits VIIa and Xa  less thrombin plasma; less efficient.
production.  Thrombin: once formed, it can cause increase in
b. PROPAGATION (platelets)  95% production.
lIa/Thrombin:
- Self-perpetuating /autocatalytic.
THROMBIN FEEDBACK MECHANISM
- When complexes are increased in phospholipids,
the more thrombin production.
ACTIVATION OF COAGULATION
NOTE: 1. Autocatalytic or self-perpetuating: many more
thrombin from small amount present
2. LOW level of thrombin activates V and VIII
3. Activates XIII  XIIIa; calcium dependent
4. Induces platelet aggregation; agonist
5. Also activates factor XI; current concept
6. Procoagulant: Activates TAFI (thrombin
activatable fibrinolysis inhibitor) – inhibits
fibrinolysis; produced by endothelial cell
The key initiating step is the exposure of TF to the
circulation and reaction of TF with factor VII. The TF- INHIBITOR TO COAGULATION
factor/VIIa complex can enzymatically activate 1. Increased thrombin: destroys Va and VIIIa  prevents
factors X and IX. The initial activation of factor X to Xa clotting/continuous clotting Disseminated coagulation
may be important in getting the coagulation cascade 2. Activate Protein C (w/ Ca++ and thrombomodulin) 
started, however, tissue factor pathway inhibitor inhibitor of coagulation (CHON S)  plasmin generation
(TFPI) rapidly inactivates the TF-VIIa-Xa complex. The  inactivation of Va Vi and VIIIa  VIIIi
major action of the TF-VIIa complex in vivo is the 3. Promote plasmin generation (fibrinolysis)
activation of factor IX to IXa, which then activates  Controls excessive coagulation
factor X to Xa. Consequently, prothrombin is  Increase concentration of thrombin (HIGH level);
converted to thrombin, then fibrinogen is converted destroys V and VII; activated protein C
to fibrin. Factors Xa and IIa have positive feedback  Protein C and S increase plasminogen activation
activity on earlier steps of the cascade.
NOTE:
HEMATOLOGY 2

- Plasmin: not normally present in plasma and it is


derived from plasminogen, which is produced by
the liver.
- Plasminogen: normally present in plasma;
precursor synthesized by liver; it will convert into
plasmin by plasminogen activators (3 activation
phases).

EXCESSIVE/ UNREGULATED FIBRINOLYSIS:

- May lead to disorders: inability to clot/excessive


clotting.

REGULATE FIBRINOLYSIS: REGULATORY PROTEINS


QUESTIONS TO REMEMBER:  PLASMINOGEN ACTIVATOR INHIBITOR (PAI)
1. What may activate II: PROTHROMBINASE: II  (Xa- - It inhibits the activators of plasminogen to
Va-Ca-PL)  IIa prevent excessive production of plasmin.
2. What may activate XII: in vivo: collagen or any
 ALPHA-2-ANTIPLASMIN
other negative surface or chemicals; in vitro: glass
- Inhibit plasmin; prevent excessive function of
contact
3. What may activate X: X  (IXa-VIIIa-Ca-PL)  Xa plasmin
4. What may activate V and VIII: THROMBIN (IIa): V FIBRIN DEGRADATION PRODUCTS (FDP) OR FIBRIN
(thrombin)  Va ; VIII  (thrombin)  VIIIa SPLIT PRODUCTS (FSP)
5. What may activate XI: XI  (XIIa, IIa)  XIa for
propagation - released after the fibrin clot is broken down
6. What may activate IX: Extrinsic tenase and XIa: IX
 (VIIa/TF) or (XIa)  IXa ACTIVATION OF PLASMINOGEN → PLASMIN
7. What activation reactions can be by-passed: XII  WAYS OR PHASES: PLASMINOGEN ACTIVATORS
XIIa (even without, coagulation can still occur)
1. INTRINSIC ACTIVATION: XIIa AND KALLIKREIN
- by XIIa-Kallikrein-HMWK
- Contact factors are closely related to the
FIBRINOLYSIS fibrinolytic system: → XIIa-Kal → plasminogen
- Last phase of Coagulation  plasmin
- Re-establishment of blood flow → lysis of clot.
- It is defined as the dissolution of clot or lysis of
clot by plasmin or fibrinolysin.
HEMATOLOGY 2

2. EXTRINSIC ACTIVATION: TPA (tissue type) 2. Fragment X will be acted on by plasmin


- Tissue Plasminogen Activator(TPA): produced by asymmetrically (one side) to separate one of
endothelial cells the D’s  Fragment Y (E-D combination)

3. EXOGENOUS ACTIVATION 3. Plasmin will act on Fragment Y and separate E


- introduced into patient: Urokinase and from the D.
Streptokinase Therapeutic and fibrinolytic agent:
prevent occurrence of abnormal clot FIBRINOGEN DEGRADATION PRODUCTS (FDP)
o Urokinase: present in the urine
 X and Y: early degradation products
o Streptokinase: exogenous; comes from
strep organisms  D and E: late degradation products

INHIBITORS NOTE:
 Plasminogen: Histidine-rich glycoprotein Fibrinogen Degradation Product = XYDE
 Activators (TPA): Thrombin Activatable
Fibrinolysis Inhibitor(TAFI) & Plasminogen
FIBRINOLYSIS
Activator Inhibitor (PAI)
FIBRIN
 Plasmin: plasmin inhibitor (alpha-2-antiplasmin;
alpha-2-macroglobulin) - Fibrin monomers (connect end-to-end (D-D
connection) and side-by-side (D-E-D connection)
FIBRINOGEN LYSIS
 fibrin polymerization XIIa (stabilizes clot;
FIBRINOGEN
crosslinks by forming covalent linkages between
- Made up of three polypeptide chains: alpha, D-D).
beta, and gamma.
FIBRIN DEGRADATION PRODUCTS X, Y, D-D DIMER, E
ALPHA CHAIN - Acted on by Plasmin

- first site that plasmin will be acted on: D-DIMER/ D-D PRODUCT
1. Plasmin will cleave alpha chains  Fragment X
(D-E-D) - indication (specific) of fibrin lysis (clot formed)
- Fragment X: first fragment to be formed; - main difference between the products
largest.
YY COMPLEX/ YY FRAGMENT: E-D-D-E

DXD COMPLEX: D-D-E-D-D


HEMATOLOGY 2

DIMER: clot Fibrinolysis is activated within 24-48 hours.

MONOMER (FIBRINOGEN): no clot formed COAGULATION MECHANISM


Coagulation and Fibrinolysis should have balance
Excessive coagulation → thrombosis
Excessive fibrinolysis → bleeding

NOTE:
 Fibrin Degradation Product: X Y D (D=D; D-
dimer) E
 In normal coagulation/hemostasis/fibrinolysis
→ X, Y, D (D-D Dimer), E will be cleared by the
liver but not in excessive fibrinolysis and liver
diseases; or if increase production of FDP → NATURALLY OCCURRING INHIBITORS OF
bleeding. COAGULATION AND FIBRINOLYSIS
SERPIN FAMILY (Serine Protease Inhibitors)

EFFECTS OF FDP: INHIBIT OR INTERFERE IN NORMAL SERINE PROTEASES


COAGULATION - Those when activated there function is to
 X and Y: with anticoagulant effects activate another enzyme in the cascade.
 Y and D: inhibit fibrin polymerization - e.g. VII → VIIa → IXa
 Fragment E: inhibits thrombin
NAME FUNCTION
NOTE: Tissue factor pathway With Xa, binds TF:VIIa
Primary Hemostasis: rapid in response; platelet plug: inhbitor
short lived and unstable Thrombomodulin Endothelial cell surface
Secondary Hemostasis: slower response; fibrin clot: receptor from thrombin
long live and stable Protein C Serine protease
Protein S Cofactor
SUMMARY OF STEPS IN HEMOSTASIS Antithrombin Serpin
1. Vessel injury Heparin Cofactor II Serpin
2. Vasospasm (TxA2; serotonin) Z-dependent protease Serpin
3. Platelet plug → PRIMARY HEMOSTASIS inhibitor
4. Insoluble fibrin clot
Alpha 1 -protease Serpin
5. Clot retraction
inhibitor (alpha1 –
6. Clot dissolution or fibrinolysis
antitrypsin)
Alpha2-macroglobulin Serpin
Clot formation (in vivo)= 6 minutes
Clot formation (in vitro) = Red tube (15-20 minutes →
1 hour completely clotted)
HEMATOLOGY 2

INHIBITOR COAGULATION FIBRINOLYSIS NOTES


Component(s) Component(s)
inhibited inhibited
Antithrombin III Thrombin, VIIa, IXa, Plasmin  First inhibitor discovered and tested
Xa, XIa and XIIa and  Main inhibitor of the entire coagulation
Kallikrein mechanism; major inhibitor of thrombin
 Enhanced by a co-factor Heparin
 It is a serpin.

THROMBIN FUNCTIONS:
1. Induces platelet aggregation
2. Activates factors XIII, V, VIII, XI
HEPARIN: does not remove Ca++

ROLE of HEPARIN: Cofactor of AT-III  inhibit


thrombin; serves as a bridge. Heparin when
bound to AT-III causes a morphological change to
be more specific to the size of thrombin.
Thrombomodulin Thrombin  It binds to thrombin and inhibits
thrombin (functions to activate protein
C).
 Produced by endothelial lining and it is a
membrane bound.
 Thrombin when bound to
thrombomodulin can’t activate factor VIII
and V instead it will activate protein C to
become Activated Protein C (APC).
 Trimolecular complex: Thrombomodulin
+ thrombin + Protein C + (receptor:
EPCR)  APC.
Thrombin INHIBIT
Activatable FIBRINOLYSIS
Fibrinolysis
Inhibitor(TAFI)

Plasminogen
Activator Inhibitor
(PAI)
Tissue Factor Xa & TF:VIIa  Kunitz-type serine protease inhibitor
Pathway Inhibitor complex that has a double inhibitory effect
(TFPI)  Major inhibitor of extrinsic pathway
3 Kunitz type:
Lipoprotein Kunitz 1: binds with VIIa; inactivating therefore
Associated VIIa-TF complex
Coagulation Kunitz 2: binds with Xa; inactivating Xa
Inhibitor (LACI) Kunitz 3: unknown fxn

Extrinsic Pathway X (intrinsic tenase: IXa-VIIIa-Ca-PL) or (extrinsic


Inhibitor (EPI) tenase: TF-VIIa) Inhibited by TFPI  Xa Inhibited
HEMATOLOGY 2

by TFPI  NO: CAN’T HAPPEN: II 


(prothrombinase)  IIa

Alpha-2 Thrombin & Plasmin  Inhibits plasmin when alpha-2


macroglobulin Kallikrein antiplasmin is depleted.
C’1 inactivator/ XIIa, XIa,& Plasmin  Inhibitor in the complement pathway
C’1 esterase Kallikrein
inhibitor
Protein C and S Va and VIIIa Enhances plasmin  Cofactor: Protein S
system formation   Vit. K dependent inhibitor
ENHANCES  Inactivates inhibitors of plasminogen
FIBRINOLYSIS activators
 Produce as zymogen by the liver
 Activated by thrombin
 APC will inactivate VIIIa and Va  VIIIi
and Vi
Protein S: exists in 2 forms in plasma
Bound to C4bBP(complement pathway;
inflammation): 60%
Free Protein S: 40%  can ONLY serve as
cofactor
In inflammation (excessive coagulation): 85-90%
bound and 10-15% is free: can’t inactivate VIII
and V.
Alpha-1- XIa Plasmin  Inhibits plasmin only when a-2
antitryspin/ A-1 antiplasmin and a-2 macroglobulin are
protease inhibitor depleted.
Alpha-2- Plasmin  Major inhibitor of plasmin
antiplasmin  First to inhibit plasmin
Z-dependent Xa, XIa  Enhanced by its co-factor protein Z
protease inhibitor (CHON –Z) additional protein that is vit. K
(ZPI) – dependent like II, VII, IX, X
 Serpin – inhibits serine protease

Protein C inhibitor Thrombin, APC, TPA, urokinase  Anticoagulant


thrombin-  Pro-coagulant
thrombomodulin  Fibrinolytic inhibitor
complex

QUESTIONS ASKED:
1. Factors in the intrinsic pathway: VII, IX, XI, XII, HK, PK (deficiency in any prolongs APTT)
2. What are the cofactors: TF-VIIa; Va-Xa; VIIIa-IXa; HK-XIIa
3. The only transaminase/transglutaminase among the coagulation factor: XIII
4. What stabilizes fibrin: XIIIa
5. Vitamin K-dependent: II, VII, IX, X, C, S, Z
V- (labile factor) Pro-accelarin / acceleratorgobulin
VII- (stable factor) Pro-convertin / plasma conversion
IX- Plasma thromboplastin component = anti hemolytic factor B
XI- Plasma thromboplastin antecedent = anti hemolytic factor C
HEMATOLOGY 2

Coagulation Group Pathway Test - One reagent in APTT/PT is calcium, therefore the
factor involved prolonged excessive citrate in blood sample will bind with
If factor is the reagent calcium  prolonged test result;
deficient calcium is inactivated by excessive citrate.
(PT/APPT/
BOTH) 2. Clots in specimen
Fletcher Contact Intrinsic APTT - Clotting happens due to consumption
factor (PK) group
coagulation factors (I, V, VII, XIII).
Accelerator Fibrinogen Common Both
- REJECT SAMPLE even the clots are micro clots
Globulin (V) group
- In coagulation studies, presence of even micro
Antihemophili Fibrinogen Intrinsic APTT
c factor A clots would cause the rejection of sample.
(VIII:C)
Proconvertin Prothrom Extrinsic PTT 3. Hemolysis and Traumatic phlebotomy
(VII) bin - Tissue Factor introduction
Plasma Contact Intrinsic APTT - Hemolyzed RBC: exerts thromboplastin-like
thromboplasti activity
n antecedent - Excessive probing  damage tissue 
(XI) thromboplastin: TF:VIIa  coagulation
Plasma Prothrom Intrinsic APTT mechanism.
thromboplasti bin
n component; 4. Icterus; Lipemia
Christmas - Affects spectrophotometry: more light
factor (IX)
absorbed  concentrated.
Stuart-Prower Prothrom Common BOTH
factor (X) bin COLLECTION OF SAMPLE
1. NEEDLE GAUGE
 Adult (<25ml): G20 - 21; 1- 1.25 in.
LABORATORY EVALUATION OF COAGULATION AND
FIBRINOLYSIS  Adult (>25 mL): G19; 1 or 1.25 in.
- All coagulation testing critically depends on the  G23: Child or adult with friable, or hardened
quality of the specimen. Minimum tissue veins.
trauma and the avoidance of hemolysis are  G20, 21, or 23: Syringe with winged-needle set
essential. It is therefore important to adhere to o The smaller the bore, the slower the
principles of proper specimen collection, blood will flow into the needle
handling and preparation. o The larger the bore, the easier will blood
will flow  prevent HEMOLYSIS.
GENERAL INSTRUCTION FOR COAGULATION TESTS o For G23(smaller): Avoid hemolysis by
- Time is important: Coagulation test like in XIII gently pulling of the plunger/ gentle
assay; any factors that may affect time should be aspiration
avoided. 2. SYRINGE
- Plastic syringe or silicone-coated
SOURCES OF ERRORS AFFECTING COAGULATION TESTS: - AVOID using GLASS in all procedure: use plastic
1. Blood volume is inadequate causing excess test tube, pipettes and syringes
anticoagulant - Glass will activate XII (glass factor)
- Recommended ratio: 1:9 using Na citrate
- In routine hematology: half full is accepted 3. ANTICOAGULANT
- 3.2% sodium citrate
- In coagulation studies: tubes should be at least
90% full.
HEMATOLOGY 2

- If patient’s Hct is more than or equal to 55%,  Aspirate always using plastic
adjust the volume of citrate since excess citrate pipette.
will bind with reagent calcium.  Platelets are found in the buffy
- ADJUSTING THE VOLUME OF CITRATE: coat layer.
o C= (0.00185) (V)(100-H)
NOTE:
o Where:
Platelet Poor Plasma (PPP) preparation (<10,000/uL)
 V = blood volume in mL
 Centrifugation: 2000 xg for 10 minutes;
 H = patient’s Hct
Rodak’s: 1500 g for 15 minutes
 C= volume (mL) of anticoagulant
needed.  Collect only the upper 3⁄4 of the plasma
layer.
4. TEMPERATURE  Test immediately because exposure to air
- Most of the tests are performed at 37°C changes the pH due to loss of carbon dioxide
- Perform: STAT (↑pH).
- But if there is delay  Freeze the PPP only; DO  Store plasma at 4°C but not to exceed 2
NOT FREEZE WHOLE BLOOD/ BLOOD WITH RBC. hours or rapid freezing to -20°C.
- Storage:
o PT (extrinsic pathway: VII (stable Platelet Rich Plasma (PRP) Preparation (200,000 –
factor)): uncentrifuge (with Na citrate) 300,000/uL)
and well capped at room temperature  Centrifugation: 60 – 100 xg for 10 minutes at
for 24 hours RT; Rodak’s 50 g for 30 mins
o Never refrigerate samples for PT: cold  Separate upper portion of the sample
temp  cold activation: VII and XI
o APTT/PTT (intrinsic pathway factors:
VIII, IX, XI, XII, HK, PK): uncentrifuge for NOTE:
4 hours at 4°C. Coagulation factors are in plasma.
o Factor VIII (labile at room and ref. temp.) 2 concentration of sodium citrate:
note also V (labile factor; common 3.2% SODIUM CITRATE (recommended by NCLSI)
pathway).  V and VII (preserve well)
o Never leave at RT: VIII will deteriorate  Less affected by polycythemia vera
o Test to monitor unfractionated heparin  Normal Ratio: 1 (citrate): 9 (blood)
therapy is APTT: heparin+antiheparin In px with high hct: adjust volume of citrate to prevent
factor (PF4) excessive citrate which will bind to reagent calcium.
o Unfractionated heparin (PTT) 
separate/centrifuge in 1 hour  Platelet 3.8% SODIUM CITRATE
 easily affected by high hct samples or polycytemia
poor plasma (PPP) to prevent the action
vera.
of PF4 against the heparin.
o PPP: at 20°C for 2 weeks; at -70°C for 6 DO NOT use EDTA (labile factors are not well preserve,
inhibit thrombin-fibrinogen interaction, affects calcium)
months.
and HEPARIN (inhibits thrombin).
SAMPLE PREPARATION

- Sample Used:
o Whole Blood
o Plasma
 Anticoagulant used: 3.2%
Sodium Citrate
HEMATOLOGY 2

TEST FOR INTRINSIC AND COMMON PATHWAYS


FACTORS TO DETECT: VII, IX, XI, XII, HMWK, PK

CONTACT FACTORS: I, II, V, X

TISSUE FACTOR: where activation starts in vivo.

1. COAGULATION TIME (Whole Blood)


- All of the components needed for clotting are
present in the blood.
- When blood is placed in a glass test tube or glass
slide, the blood will be activated.
NOTE: - Factor XII is activated first by contact with glass
ROUTINE SCREENING TEST (PT/APPT): Light blue first slide (PK to Kalikrein; XIIa activate XI to XIa; Xia
SPECIFIC FACTOR ASSAY (like VIII, VII assay): use a activates IX to IXa).
prime/dummy tube, red top, light blue - Activation starts with contact activation phase
FOR OPEN SYSTEM: Two-syringe method: after - Measures the time required for whole blood to
collecting blood using the first syringe, detach clot after it has been removed from the body
syringe without withdrawing the needle and replace - Affected by amount of blood used.
with another syringe. The blood in the first syringe
will be discarded and the blood in second syringe is
A. SLIDE METHOD
the one be used.
 REFERENCE VALUE: 2-4 minutes
 REQUIREMENT: glass slide method
RULE:
1st: DISCARD B. CAPILLARY METHOD (Date and Laidlaws
2nd: USE Method)
 REFERENCE VALUE: 2-4 minutes
 REQUIREMENT: Non-heparinized
NOTE: capillary tube (blue ring) or else no
What factors will deteriorate at room temperature? V clotting will occur.
and VIII
Refrigerator temperature affects what factors? VII PROCEDURE FOR BOTH (SLIDE AND CAPILLARY
and XI METHOD):
PT plasma samples capped, are stable for 24 hours at
what temperature? Room temperature a. Fill the non-anticoagulated capillary tube & let it
APTT samples are stable for 4 hours if stored at 4°C. stand for 2 minutes
- NOTE: do not wipe off the first drop of blood but
include it.
COAGULATION TESTS b. Start time as soon as blood starts to appear from
- All based on the time it takes for the blood
the site of puncture
samples to form into a clot whether plasma or
c. For the slide method place 2-3 drops of blood in
whole blood.
a slide while for capillary tube, fill it with whole
- Accuracy in measuring time is necessary because
blood
little excess may be reported as falsely
d. Wait for 2 minutes initial time before checking
prolonged instead of normal.
for fibrin clot formation
e. CHECKING:
o SLIDE METHOD: check by using the same
lancet from collection by pricking into
the blood sample and raising the blood
HEMATOLOGY 2

sample until a thin fibrin thread is l. After 30 secs. Check for clotting for tube 2 and
observed as thin as the hair & record the same procedure will be employed as with tube
time when the thin fibrin thread is 1
observed as the clotting time m. Once clotting is complete in tube 2, wait for
o CAPILLARY METHOD: Break the glass another 30 secs. Before reading tube 3
tube on one side without pulling them n. Once 30 secs is reach, check for clotting with
apart and slowly pull them apart and tube employing the same procedure with tube
observed for a thin fibrin thread and report the clotting time
connection, connecting both fragments.
NOTE:
f. If no fibrin thread is observed wait for another
TIME DIFFERENCE BETWEEN TUBES: 30 SECONDS
30 seconds and check again and if it is not yet
REPORTING: from time the blood starts to appear until
visible again continue waiting for another 30
complete clotting time in tube 3.
seconds and check again. REFERENCE VALUE: 7- 15 MINUTES
NOTE:
BOTH NORMAL VALUE: 2-4 minutes at RT 2. ACTIVATING CLOTTING TIME (reliable)
AFFECTED BY: amount of blood used - PRINCIPLE: Whole blood contains all the
Micromethod requires fewer blood sample while for
components necessary to produce a clot when
macromethod, the longer is the clotting time.
put into a glass tube. By adding an activator (e.g.
Diatomite) and keeping blood at constant 37°C.
C. LEE AND WHITE METHOD (Macromethod) - The test becomes more reliable and rapid.
o REFERENCE VALUE: 7-15 minutes - REFERENCE VALUE: 75-120 seconds
o DISADVANTAGE: Longer clotting - ACTIVATOR: Diatomite – shortens clotting time
time due to usage of larger blood and enhances the coagulation factors
volume.

PROCEDURE (LEE AND WHITE METHOD):


NOTE:
a. Place 1 mL of each into 3 tubes with equal size
PLASMA CLOTTING TIME
and dimension (12 x 75) - Coagulation factors are in the plasma
b. Label the tube as 1, 2 and 3 - Plasma is the one which clots
c. Extract 4 mL of whole blood - Plasma clotting is translucent
d. Start time as soon as blood appears at the
puncture site.
e. Place 1 mL of blood in each tube starting from
the 3rd tube up to the first
f. Discard the last remaining 1 mL
g. Stoppered the three tubes and incubate at 37°C 1. No clot
in a water bath for 5 minutes undisturbed 2. Clotted
h. After 5 minutes, check for clot formation by 3. Clotted
tilting the tubes at 45° angle 4. Clotted
i. observed first the tube 1 up to tube 3
j. If blood flows to the tube, it is not yet fully 3. PLASMA RECALCIFICATION TIME
clotted thus incubate further for 30 sec. - Plasma was prepared using Na Citrate thus Ca is
k. After 30 secs, check tube 1 again and when removed.
tube 1 does not spill its content upon total - Plasma is calcium free thus add calcium (reagent)
inversion, clotting time is complete and record - Upon addition of calcium take the time it takes
- NOTE: do not report clotting time in tube 1 for plasma to clot
HEMATOLOGY 2

- Reference value depends on the plasma used - PRINCIPLE: Expect for Ca and Platelet
(platelet-rich plasma or platelet poor plasma) phospholipid (PPL), PPP contains all the
- No longer performed coagulation factors needed for the generation
- PRINCIPLE: except for Ca, normal PRP/PPP of intrinsic prothrombinase. When Ca is added
contains all the components necessary for with “incomplete” thromboplastin (serves as
generating fibrin. platelet substitute), intrinsic prothrombinase is
- REFERENCE VALUE: generated.
o PRP: 100-150 seconds - Difference:
o PPP: 130-240 seconds o APTT: w/ activator
o PTT: w/out activator
4. ACTIVATED PARTIAL THROMBOPLASTIN TIME
NOTE: APTT
(APTT)
FACTORS THAT MAY PROLONG THE TEST:
- Routine screening test
A. Any factor deficiency (Intrinsic (VIII, IX, XI, XII,
- Thromboplastin/Tissue Factor is involved in the
HMWK, PK) and common pathway (I, II, V, X))
extrinsic pathway which activates VII to VIIa B. Presence of inhibitor – Factors VIII-I and FIX-I
and binds to it C. Heparin (immediately process the sample and
- Test for Heparin therapy prepare the plasma within 1 hour to prevent PF4 from
- REAGENT: partial thromboplastin: cephalin inhibiting the heparin)
(phospholipid only) & 0.025 M Ca (reaction D. <60 mg/dL fibrinogen – Factor 1 belongs to
initiation common pathway.
- SAMPLE: Platelet Poor Plasma (PPP) (Hct <55%) E. Presence of increased Fibrin Split Products (FSP) –
to avoid inappropriate with citrate and plasma all clot-based test will be prolonged in the presence of
volume. FSP.
- ACTIVATORS: Kaolin, Celite, Ellagic acid NORMAL VALUE: 25 – 35 SECONDS
- PURPOSE: for determination of deficient in
intrinsic pathway
- PROCEDURE: Starts with the activation of FXII
NOTE:
upon contact with a negative surface. In vivo, it
 All of clot-based test cannot measure Factor
may be collagen while for in vitro it is the glass XIII deficiency & FXIII will be normal because it
test tube. Coagulation cascade will not proceed is not needed for formation of clot.
if intrinsic tenase and prothrombinase will not  When thrombin acts on XIII, it will now
be formed thus no fibrin formation. Both stabilize the clot.
intrinsic tenase and prothrombinase requires  XIII is involved in common pathway.
Ca & phospholipid. The source of phospholipid
source is the platelets but since the sample is
platelet poor plasma, there would be no source TEST FOR EXTRINSIC AND COMMON PATHWAYS
of phospholipid. While, Ca is removed by EXTRINSIC: VII
citrate. Thus, cephalin and Ca is added in order COMMON PATHWAY: I, II, V, X
to observe clotting. Upon addition of cephalin
and Ca, observed the time it takes to form fibrin 1. PROTHROMBIN TIME (PT) / QUICK’S TEST
clot - Starts with the action of VII.
- Most useful procedure for routine screening of - Dominant pathway that occurs in vivo, once
coagulation disorders in the intrinsic system, for thromboplastin is introduced in the circulation,
detecting the presence of circulating anti- it immediately combines with VIIa already
coagulant and for monitoring heparin therapy. present in circulation in vivo but in vitro, VII
Also measures factors present in the common must be activated by thromboplastin. In vitro,
pathway, except for platelet and factor XIII. there is no source of thromboplastin when
HEMATOLOGY 2

placed in a test tube. The reagent  Oral anticoagulants (warfarin, coumarin,


thromboplastin will provide the thromboplastin coumadin).
which comes from tissues. Complete
thromboplastin is needed because it has to SOURCES OF ERRORS:
supply the other missing factors such as  Prolonged in the presence of heparin
phospholipid and thromboplastin. The contamination; can be shortened in traumatic
composition of complete thromboplastin is venipuncture.
REPORTING:
tissue factor and phospholipid. The tissue factor
 INR to monitor effect of therapy and dose of
in the reagent will activate VII and the
oral anticoagulant
phospholipid in the reagent will supply the
ADDITIONAL REMINDER:
missing phospholipid. Since citrate is utilized, Ca
 Prolonged PT causes VII to disappear
is also removed thus 0.025 M CaCl2 is utilized immediately since it has a shortest lifespan in
as a substitute. Upon addition of vivo.
Thromboplastin and CaCl2, record the time it
takes for plasma to form into clots
- Useful screening procedure for the extrinsic INTERNATIONAL NORMALIZED RATIO (INR)
coagulation mechanism including the common - A standardized way of reporting PT in
pathway, also monitors oral anticoagulant monitoring oral anticoagulant therapy,
therapy with coumadin. calculated as a ratio of the patient’s PT to a
NOTE: PT control PT standardized for the potency of the
 PRINCIPLE: when tissue extract or thromboplastin reagent developed by the
thromboplastin is added to PPP along with Ca, World Health Organization (WHO).
it reacts with factor VII leading to subsequent - Reference Values for INR:
activation of factor X. o 2.0-3.0
 NORMAL VALUE: 10 – 12 SECONDS  In prevention and treatment of
 THROMBOPLASTIN SOURCE: rabbit brain venous thrombosis
extract or similar  Treatment of pulmonary
 SPECIMEN: Citrated PPP to prevent excessive embolism
citrate in plasma  Prevention of stroke in MI
 REAGENTS: Complete Thromboplastin (TF &  Peripheral arterial disease
phospholipid) and 0.025 M CaCL2  Prevention of systemic
 PURPOSE: in monitoring extrinsic and embolism in atrial fibrillation
common pathway deficiency, more sensitive
 Cardiac value replacement
for monitoring oral anticoagulant therapy,
(tissue valves).
vitamin K deficiency and liver diseases

o 2.5-3.5
NOTE:  In prevention of recurrent MI
PROLONGED PT IS SEEN IN:  Reduction of mortality in MI
 Any factor deficiency under the extrinsic (VII,  Mechanical prosthetic heart
I, II, V, X) and common pathway. valve (high risk).
 Fibrinogen concentration <100mg/dl
 Dysfibrinogenemia – abnormal fibrin FORMULA:
𝑝𝑎𝑡𝑖𝑒𝑛𝑡 𝑃𝑇
structure affecting PT. INR= [ ]𝐼𝑆𝐼
𝑃𝑇 (𝑐𝑜𝑛𝑡𝑟𝑜𝑙 𝑚𝑒𝑎𝑛)
 Vitamin K deficiency (II, VII, IX, X) and certain
liver disease (affecting all factors except, VWF,
Ca & TF synthesis) thus prolong PT and APTT
HEMATOLOGY 2

SURFACE ACTIVATION – initiates intrinsic pathway Perform


when factor XII is in contact with any surface; mixing
monitored by APTT. studies

TISSUE THROMBOPLASTIN
2. STYPVEN TIME/ RUSSEL VIPER VENOM TEST
- for extrinsic and monitored by PT - Uses a venom (from (Viperarusselli/Daboia
INTRINSIC PATHWAY – PTT russelli) with a “thromboplastin like” action
- Principle: Addition of the venom bypasses the
VII – PT activation of VII and directly activates factor X
INTRINSIC PATHWAY AND FACTOR VII – common - Reagent: Russel Viper Venom 
pathway; APTT & PT Thromboplastin-like (“VIIa”): VIIa like activity 
immediately activate X.
- Activation of VII is bypassed in the reaction
because reagent have VIIa-like activity
- Initial utilization: Differentiate X and VII
deficiency
o X deficiency: ST: Prolonged
o VII deficiency: ST: Normal
- Clinical utility now: for identification or detect
the presence of Lupus anticoagulant (LA) or
Anti-phospholipid Antibody (APA) – antibody
against phospholipid; inhibits PF3 and
phospholipid (interferes normal coagulation)
prolonged test.
- Screening test performed using diluted plasma:
Dilute Russel Viper Venom Test (DRVVT) or
Dilute Stypven Time (DST) for Lupus
Anticoagulant: factor activity INCREASES with
increasing dilutions of patient plasma used in the
assay (Corresponding dilutions of LA/APA).
- Performed in combination with APTT: ST and
APTT  identify LA/APA.
TEST INTRINSIC EXTRINSIC COMMON - Interpretation:
PT- None Prolonged None o CF deficiency (dilution) : more
prolonged Factor VII prolonged
APTT- deficiency o LA-causing (diluted): improved in the
normal test/ shorten
APTT- Prolonged None None  Rationale: while the plasma is
prolonged VIII, IX, XI, being diluted, the LA is also
PT- Normal XII, HMWK, diluted  increase CF activity
PK - Circulating anticoagulant: abnormal
deficiency
anticoagulants that inhibit certain coagulation
Perform
proteins.
additional
testing - Specific inhibitor: inhibitor/anticoagulant that
BOTH PT & None None Prolonged inhibits specific coagulation protein like factor
APTT- I, II, V, X VIII or IX
prolonged
HEMATOLOGY 2

- Non-specific inhibitor/ non-specific circulating o Normally prolonged in the newborn and


anticoagulant: inhibitor/anticoagulant that in multiple myeloma.
inhibits no specific coagulation protein like LA.

INDIRECT TESTS:
FOR FIBRINOGEN DEFICIENCY ONLY
THROMBIN TIME (FIBRINOGEN DEFICIENCY TEST)

- Modification of Thrombin Clotting Time (TCT) by


Clauss.
- Principle: Addition of thrombin bypasses all
coagulation pathways except the polymerization
of Fibrinogen.
- Specimen: citrated PPP APTT: intravenous anticoagulant
- Reagent: standardized thrombin solution and
calcium. PT: oral anticoagulant  INR
o Fibrinogen --- Standard Thrombin + Ca++ TT: fibrinogen deficiency and heparin therapy
 Fibrin
- Will not detect deficiency in the CF VII, V, X, VIII, TT together with APTT: Heparin therapy
IX, XI, XII, HK, PK  reactions bypassed
DST together with APTT: LA
- Cannot determine deficiency or availability of
thrombin. REPTILASE TIME
- DETECT FIBRINOGEN DEFICIENCY ONLY - Uses the reagent reptilase enzyme (collected
- Reactions involved conversion of fibrinogen to from Bothropsatrox) which has a thrombin-like
fibrin. effect.
- Measures the availability of adequate and - Functionality same as thrombin time: IDENTIFY
functional fibrinogen PRESENCE, AVAILABILITY AND FUNCTIONALITY
- Used to monitor heparin therapy and is sensitive OF FIBRINOGEN.
to presence of increased levels of FSP. - REAGENT: Reptilase (“IIa” – like): toxic and
- Presence of FSP: XYDE; inhibits fibrin potent: very unsafe to handle (should not be in
polymerization and inhibits conversion of contact in open skin  blood will clot)
fibrinogen to fibrin by thrombin  inhibits - Fibrinogen --- Reptilase  Fibrin ; immediate
clotting/prevent clot formation  increased/ clotting will occur
prolonged TCT (more sensitive). - PRINCIPLE: Reptilase bypasses all coagulation
- Monitors Heparin Therapy pathways except the polymerization of
o Heparin: anti-thrombin; only fibrinogen.
anticoagulant (in vivo and in vitro) that - FIBRINOGEN: 3 polypeptide chains (alpha, beta
does not remove Ca++; functions to and gamma) ends with D-terminals and a central
inhibit thrombin by increasing the E domain where alpha, beta and gamma
activity of AT-III. polypeptide chains meet and coil extended at E
o APTT and TT: monitors heparin therapy central domain is fibrinopeptides A and B.
- Reference value: 17-25 secs. (may depend on - If thrombin cleaves fibrinogen: thrombin
the thrombin concentration) cleaves both fibrinopeptides A and B.
- Prolonged Thrombin time: - If reptilase cleaves fibrinogen: reptilase cleaves
o Fibrinogen level is below 75-100 mg/dL; only fibrinopeptides A.
functional disorder of fibrinogen - Effect are similar: Fibrinogen  fibrin monomer
o Presence of Heparin, fibrin degradation  polymerization
products (FDP) and thrombolytic agents
HEMATOLOGY 2

- Reptilase cleaves I  monomers polymerize F XIII Deficiency (soluble Dissolved Not


(end-to-end)  formation of clot clot) dissolved
o Thrombin: end to end and side to side Excessive fibrinolysis Dissolved Dissolved
connection (excessive plasmin)
o Reptilase: end to end only
- Prolonged reptilase time is observed in OTHER TEST:
Fibrinogen deficiency and other functional
disorder of fibrinogen; presence of FSP/FDP 1. BETHESDA ASSAY: determine and measure the
- REFERENCE TIME: 18-20 secs titer of specific factor inhibitor of VIII.
- More sensitive to fibrinogen deficiency 2. ECARIN CLOTTING TIME

NOTE: HISTORICAL TESTS:


 All coagulation assay is affected by the Basis of coagulation tests
presence of FSP  prolonged PROTHROMBIN CONSUMPTION TEST
 BOTH (Fibrinogen Assay) are used to identify
- Deficiency in factors V, VIII, IX, X, XII or platelets
availability and functionality of fibrinogen and
will slow the rate of prothrombin conversion
both sensitive to fibrinogen deficiency or
dysfunctional  prolonged test leaving significant amount of prothrombin in the
serum.
- Prothrombin Time (PT) is performed on SERUM
THROMBIN REPTILASE - Deficiency in some factors will SLOW the rate of
TIME (standard TIME prothrombin conversion leaving significant
thrombin Rgt) (thrombin-like amount (60%) of prothrombin in serum
rgt.) o Slow pathway: Prothrombin (60%) 
Heparin – both Prolonged Normal or
Thrombin (40%)
APTT and TT Slightly
- Can’t identify which coagulation factor is
Prolonged
deficient  needs mixing studies.
(overdosed)
(+) FSP – all Prolonged Prolonged - GENERAL RULE: Only consumable factor are
clotting test FIBRINOGEN GROUP (I, V, VIII and XIII) and
Decreased Prolonged Prolonged factor II.
Fibrinogen - During clotting (serum): II (20%; residual
prothrombin)  IIa (Thrombin: 80%)
- SERUM: product of clotting of blood
DUKERT TEST (5M UREA SOLUBILITY TEST)

- Test for Factor XIII


- PRINCIPLE: clot formed in normal plasma is
insoluble in 5M urea during a 24-hr incubation.
- If factor XIII is deficient in the patient’s plasma,
the clot is dissolved in less than 24 hours by the
urea.

1% monochloroacetic acid Incubate for 24 hours


at RT
Clot + 5M Clot+
THROMBOPLASTIN GENERATION TEST (TGT)
Urea NaCl
(NSS) - By Biggs & Douglas
Normal (insoluble clot) Not Not - Uses BaSO4/Al(OH)3- adsorbed plasma or fresh
dissolved dissolved serum
HEMATOLOGY 2

- Basis of mixing studies AUTOMATED INSTRUMENTS:


- No longer performed
 Ortho Koagulab 16S and 40A
- Mixing agent + Patient’s plasma  clotting
- Mixing agents: BaSO4/Al (OH)3 – Adsorbed  Coag-A-Mate X2 and XCm
plasma; Fresh serum; stored serum.  MLA Electra 700 and 800

HICKS-PITNEY TEST MIXING STUDY OR SUBSTITUTION STUDIES


- Performed when PTT & PT are prolonged to
- Modification of Thromboplastin Generation determine whether it is caused by a nonspecific
Time (TGT) and requires re-calcification of inhibitor, specific factor inhibitor, or coagulation
diluted plasma. factor deficiency, and to determine the deficient
coagulation factor.
AUTOMATION IN COAGULATION
- Procedure: Mix equal parts of patient’s plasma
- Detects formation of FIBRIN
plus Fresh normal plasma then repeat the
FIBRINOMETER prolonged test.

- A semi-automated mechanical instrument that RESULT AND INTERPRETATION:


detects fibrin strand formation using a wire loop
 Presence of a pathological circulating
or hook.
anticoagulant: if test is not corrected (remains
- Only machine in hematology that uses electro-
prolonged) when mixed with mixing normal
mechanical (manual) principle.
plasma.
- Fibrinometer has wells where samples are
placed, and has a wire loop or needle which dips  Factor deficiency: if repeated test is corrected
in the blood sample in the wells and keeps when mixed with normal plasma. The next thing
dipping in 0.5 seconds interval (up and down) to do is to identify the deficient factor by
until fibrin thread or clot formation is observed. performing additional mixing study or factor
When fibrin threads from the sample connects assay.
with the wire loop, stop the time and record the  Identification of deficient factor: mix equal parts
clotting time of patient’s plasma plus an appropriate mixing
- Semi-automated. sample.
- Identify fibrin formation FACTOR ASSAYS:
PHOTO-OPTICAL 1. Fibrinogen assay: Clauss modification of
- Depends on the increase in light scattering Thrombin time.
associated with the conversion of soluble  SINGLE FACTOR ASSAY:
fibrinogen molecules to the insoluble o Quantification of remaining deficient
polymerized fibrin clot. factor; determine the factor level in
- Machines measures: increase in light scattering relation to
- Identify fibrin formation from fibrinogen o Applicable only in A and B: sex linked
- In plasma, fibrinogen is dissolved thus it is not o Not for: C (autosomal)
able to scatter the light into different direction o Do dilution: 1:10; 1:20; 1:40; 1:80  CT
- Fibrinogen  Fibrin (insoluble) fibrin particles interval (Reference Curve).
will block and scatter the light in different Hemophilia: Factor Level vs. Severity
direction. Severity Factor Level Manifestation
SEMI-AUTOMATED INSTRUMENTS: Severe <1% Spontaneous
bleeding; bleeding
 Electra 750 and 750A, Fibrin timer series; and FP with minor surgery or
910 coagulation analyzer trauma
HEMATOLOGY 2

Moderate 1-5% Spontaneous bleeding Factor Deficiency Mixed Reagent Result


uncommon; may (Patient’s
bleed with surgery or sample)
trauma. VII Factor-VII No correction
Mild 5-20% No spontaneous Deficient Plasma
bleeding; may bleed
with major trauma or
surgery 4. Factor XIII assay: Chromogenic assay
Example: - Transglutaminase/Fibrinase/Transaminase
APTT: prolonged PT: Normal reaction  Release of ammonia
APTT: not corrected by Factor XI- deficient plasma but is - Directly proportional: More ammonia release:
corrected by Factor IX-deficient plasma more XIII deficiency; Less ammonia release: less
 What type of hemophilia? XIII deficiency
o Hemophilia C - (Glu DH Rxn): NADPH consumption is measured
by decrease of A at 340 nm
2. Single factor assays using PTT - Absorbance is inversely proportional to factor
- For factors VIII (A-most common), IX (B- second XIII activity.
common), and XI (C-rarest) deficiencies - Inversely proportional: High absorbance:
(hemophilias). Increase XIII deficiency; Lower absorbance:
- Uses a factor-depleted platelet-poor plasma (like Normal or Increase
factor VIII-depleted PPP which provides normal - Factor XIII: Assay cannot be detected by PT and
activity of all procoagulant except factor VIII). APTT
- Normal plasma  normal result
e.g. Px plasma + factor deficient plasma NOTE:
Prolonged Test (PT, APTT/Both coagulation test) 
APTT reagent
CONSIDER/POSSIBLE REASONS: px is suffering in
PT reagent
coagulation factor deficiency or abnormal component
FACTOR MIXED REAGENT RESULT (inhibitor/circulating anticoagulant) that keeps
DEFICIENCY inhibiting the factors  perform mixing study
(Patient’s Reagent: 1:1 mixture
- Equal parts: Patient’s plasma + correction
Sample)
reagent or mixing reagent
VIII Factor-VIII Deficient No correction
PRINCIPLE:
Plasma - If patient is deficient and we add a mixing
Factor-IX Deficient Corrected reagent that contains all necessary
Plasma components  corrected: factor deficiency.
IX Factor-VIII Deficient Corrected - If patient is deficient and we add a mixing
Plasma reagent that contains all necessary
Factor-IX Deficient No correction components but still not corrected: presence
Plasma of abnormal component.

e.g. repeat prolonged test but: (1:1) Px plasma+


3. Single factor assays using PT normal fresh plasma(contains all coagulation factor)
- For factors II, V, VII, and X deficiencies
HEMATOLOGY 2

PROLONGED TEST
INTERPRETATION NORMAL FRESH PLASMA
Factor Deficiency Normal: Corrected test
Inhibitor; C Remains Prolonged: not
anticoagulant corrected test

NOTE:
After Coagulation test (prolonged)  perform mixing
study with Fresh Normal Plasma (FNP): identify
whether it is coagulation deficiency or presence of
abnormal component  then identify which factor is
deficient or abnormal component
PROLONGED TEST: Deficiency  continue mixing
studies
PT: VII
APTT: VIII, IX, XI, XII, HK, PK
PT and APTT: I, II, V, X

MIXING STUDIES
MIXING/CORRECTION REAGENT CF PRESENT CF ABSENT
FRESH NORMAL PLASMA ALL NONE
ADSORBED PLASMA I,V, VIII, XIII, XI, XII, HK,PK II, VII, IX, X
AGED PLASMA I, XIII, II, VII, IX, X, XI,XIII, HK, PK V and VIII
FRESH SERUM II, VII, IX,X,XI,XII,HK,PK I, V, VIII, XIII
AGED SERUM VII,IX,X,XI,XII,HK,PK I, V, VIII,XIII, II (residual II
deteriorates)
ADSORBED SERUM XI, XII, HK, PK I, V, VIII, XIII, II, VII, IX, X
FACTOR DEFICIENT PLASMA Deficient of particular CF; removed through immunoadsorption
Commercially available plasma (named)
e.g. Factor VIII deficient plasma  deficient of VIII

3 WAYS COAGULATION FACTORS ARE REMOVED: EXAMPLES


PROBLEM 1: PT: Prolonged & APTT: Prolonged
1. Clotting/Coagulation: Consumable factors (I, V,
XIII) and partially II. 1. Establish whether it is coagulation deficiency or
2. Adsorption: Adsorbable factors presence of abnormal component
3. Storage/Aging: Labile factors (V and VIII) Perform test with FNP  if corrected: CF
- Aged Serum: residual prothrombin deficiency
HEMATOLOGY 2

2. Identify which pathway is affected: Common  Elimination:


pathway. Adsorbed plasma (VIII and XI present) (IX absent)
3. Coagulation Factor Deficiency (CFD) (I, II, V, X)  Fresh serum (IX and XI present) (VIII absent)
Mixing study.
ANSWER: Factor VIII deficiency
4. CFD: Pxt Plasma + Mixing Rgt.  Repeat PT and
APTT.

PROBLEM 2: COMMON PATHWAY PROBLEM 3: PT and APTT – prolonged


 Coagulation Factor Deficiency: I, II, V, X  Both are corrected by fresh serum but not with
1. Pxt Plasma + Mixing Rgt: Fresh Serum (I, V stored serum. Identify factor/s deficient.
absent; II, X present)  Repeat PT and Aptt.  Common pathway deficiency: I, II,V, X  factor II
Deficient: deficiency
I = not corrected
II = corrected A. VIII B. I C. V D. II E. X
V = not corrected  Elimination:
X = corrected o Fresh serum (II and X present) (I, V
2. Pxt Plasma + Mixing Rgt: Aged Plasma (V absent)
absent; I, II, X present)  Repeat PT and APTT o Stored serum (X present) (I, V, II absent)
Deficient:
I = corrected NOTE:
II = corrected WAYS OF ANALYZATION
V = not corrected: prolonged  ELIMINATION
X = corrected  WRITING
 RECORDING
PROBLEM 3: PT and APTT are prolonged and corrected
by addition adsorbed plasma. Which coagulation is/are
deficient? PROBLEM 4: PT and APTT – prolonged

 Pxt Plasma + Mixing Rgt: Adsorbed Plasma ( II  Both are corrected with fresh serum but not with
and X absent; I and V present)  Repeat PT and adsorbed plasma.
APTT o Fresh serum: II, VII, IX, X, XI, XII, HK, PK
 Deficient: (present)
I = corrected o Adsorbed plasma: II, VII, IX, X (absent)
II = not corrected  What may be mixed to identify the factor
V = corrected deficient?
X = not corrected o Stored serum: II and X

ANSWER: Factor I and V deficiency PROBLEM 5: PT and APTT – prolonged

PROBLEM 4: PT: Normal APTT: Prolonged  COMMON PATHWAY: I, II, V, X


 Both are NOT corrected with fresh serum but
 Corrected (it contains the CF) by adsorbed corrected with adsorbed plasma.
plasma but NOT(absent) by fresh serum Factor o Fresh serum: I, V,VIII,XIII (absent)
VIII deficiency o Adsorbed plasma: I,V, VIII, XIII (present)
 Factor deficiency in INTRINSIC PATHWAY: VIII, IX,  What may be mixed to identify the factor
XI (common deficiencies to occur); (XII, PK, HK – deficient?
deficiency will not cause bleeding disorder but o Aged Plasma: I
may cause prolongation of the test and are very
rare to occur)
HEMATOLOGY 2

PROBLEM 6: PT and APTT – prolonged

 COMMON PATHWAY: I, II, V, X


 Both are NOT corrected with fresh serum but
corrected with adsorbed plasma and aged
plasma.

FACTOR PROLONGED TEST/S CORRECTED BY NOT CORRECTED BY


DEFICIENCY
I PT and APTT  Adsorbed plasma  Fresh Serum
 Aged Plasma  Aged Serum
 Adsorbed Serum
II PT and APTT  Aged plasma  Adsorbed plasma
 Fresh serum  Aged serum
 Adsorbed serum
V PT and APTT Adsorbed plasma  Aged plasma
 Fresh serum
 Aged serum
 Adsorbed serum
VII PT  Aged plasma  Adsorbed plasma
 Fresh serum  Adsorbed serum
 Aged serum
VIII APTT Adsorbed plasma  Aged plasma
 Fresh serum
 Aged serum
 Adsorbed serum
IX APTT  Aged plasma  Adsorbed plasma
 Fresh serum  Adsorbed serum
 Aged serum
X PT and APTT  Aged plasma  Adsorbed plasma
Stypven Time  Fresh serum  Adsorbed serum
 Aged serum
XI APTT ALL NONE
Presence: APTT and Stypven Time NONE ALL
AntiPL-
Antibody
NOTE: Factor XIII Assay cannot be detected by PT and APTT
HEMATOLOGY 2

MIDTERMS - The effect of coagulation is consumption of


DISORDERS OF FIBRINOLYSIS coagulation factors (I, V, VIII and XIII) and
PLASMIN platelets. The effects of activation of fibrinolysis
is production of FSP. Main result: continuous
- Main protein involved that dissolves the clot
bleeding
and may cause excessive fibrinolysis.
- Regulatory proteins: alpha 2 antiplasmin: main
- Comes from plasminogen in the presence of
inhibitor of plasmin; and other inhibitors are
plasminogen activators like tissue plasminogen
consumed because of too much production of
activator and activated factor XII (XIIa) when
plasminogen activators.
produced can dissolve fibrin and if excess
fibrinogen. TESTS FOR FIBRINOLYSIS
EUGLOBULIN CLOT LYSIS TIME
FUNCTIONS OF PLASMIN:
- Principle: Plasma euglobulins are precipitated
 Plasmin  fibrinogen  Fragment X, Y and
with 1% HAC. Precipitates are redissolved and
Fragment D, E
clotted with thrombin. The clot is incubated, and
 Cross-linked fibrin  X-oligomers and D-dimer
the time for complete lysis at 37°C is measured.
NOTE: - Procedure: Precipitation of plasma euglobin
In both degradation and split products of fibrin or with 1% Acetic acid  re-dissolved ppt  add
fibrinogen are produced. thrombin: fibrinogen immediately converted
into fibrin  plasminogen present in fraction
now present in fibrin will be slowly converted
1. PRIMARY FIBRINOLYSIS (FIBRINOGEN LYSIS)
into plasmin by activators of plasminogen 
- This is due to the release of excessive activators
fibrin clot with plasmin  check the
of plasminogen resulting to the conversion of
ability/activity of plasmin to dissolve the clot 
plasminogen to plasmin in the absence of fibrin
at 37°C  CLOT LYSIS TIME
formation
- TEST FOR PRIMARY FIBRINOLYSIS
- Fibrinolytic system: activated in the absence of
- Detects excess production of plasmin
clot formation.
- Separation through PRECIPITATION: inhibitors
- Main problem: Increase activators of
left in the supernatant
plasminogen that leads to increase production of
- Plasma Euglobulin Fraction contains
plasmin.
Plasminogen, Activators of plasminogen and
- Since the function of plasmin is not specific to
fibrinogen.
the clot, it also destroys intact fibrinogen
- Normal clot lysis time: within 2-4 hours.
together with factors V and VIII.
- Clot lysis in less than 2 hours is indicative of
- Effect of plasmin on plasminogen: will cause the
abnormal fibrinolytic activity
release of FSP which interferes in normal
- Increase fibrinolysis is seen in: circulatory
clotting; unable to clot  BLEEDING.
collapse; adrenalin injections; sudden death;
- To inhibit the role of plasmin: treatment
pulmonary surgery; pyrogen reaction; obstetric
involves introduction of anti-fibrinolytic
complications
drugs/antiplasmin.
TESTS FOR SECONDARY FIBRINOLYSIS
2. SECONDARY FIBRINOLYSIS (FIBRIN LYSIS) - Detects fibrin monomers
- This clot dissolution results to increase FSP/FDP
ETHANOL GELATION TEST (BREEN AND TULLIS) AND
that interfere with coagulation and platelet
PROTAMINE SULFATE TEST
function.
- In DIC where there is a simultaneous occurrence - Principle: 50% ethanol (and Protamine sulfate)
of both coagulation and fibrinolysis. will cause the soluble fibrin monomers
HEMATOLOGY 2

complexes to dissociate resulting in the product came from primary or secondary


polymerization of the monomers and fibrinolysis.
subsequent gel like clot formation or - POSITIVE: presence of FSP  agglutination.
paracoagulum (paracoagulation)
PRIMARY SECONDARY
- Both tests are designed to detect the presence
FIBRINOLYSIS FIBRINOLYSIS
of fibrin monomers in the plasma. A screening
Platelet Normal Decreased
procedure utilized as an aid in the diagnosis of
count  Platelet  Platelets
DIC does not are
- Normally, there is NO gel formation. Because undergo consumed
there should be no fibrin monomers. aggregati during clot
- Presence of paracoagulation: indicates on thus is formation
presence of fibrin monomers and secondary not
fibrinolysis. consume
- Differentiate Primary and Secondary Fibrinolysis: d
o Negative: Primary fibrinolysis RBC Normal RBC
o Positive: Secondary fibrinolysis morpholog  No clot fragmentation:
y formatio Schistocytes
D-DIMER TEST/ D-D ASSAY n thus  Caused by
there is trauma
- Specific to SECONDARY FIBRINOLYSIS only no
- Measures fibrin formation (presence of D-dimer trauma
indicates: fibrin clot and fibrin lysis) caused in
- D-D fragment arises from the degradation of RBCs in
cross-linked fibrin. Its presence is specific of the
fibrin formation. circulatio
- The test uses latex beads with monoclonal Ab n
that will react only with D-dimer (Turbidimetry PT and Prolonged Prolonged
or Nephelometry) APTT  Excessive  Consumpti
- Normal: <200ng (D-dimer)/ (blood) mL : action of on of I,V,
<200ng/mL. plasmin VIII and XIII
on I, V during clot
- Increased results may be seen in DIC,
and VIII formation
thrombosis, phlebitis or other condition

characterized by fibrin clot formation. excessive
THROMBO-WELLCOTEST (TEST FOR FSP) fibrinolysi
s
- PROCEDURE: Whole blood + thrombin (for FDP’s or POSITIVE POSITIVE
complete clotting of whole blood)  serum + FSP’s
soya bean enzyme (as inhibitors)  diluted D-dimer NEGATIVE POSITIVE
serum + latex particles (coated with anti-FSP)  Euglobin POSITIVE NEGATIVE
presence of FSP  agglutination Clot Lysis
- Whole blood is added to thrombin to ensure Protamine NEGATIVE POSITIVE
complete clotting and soya bean enzyme Sulfate(SO
4) Test
inhibitor after incubation, the patient’s serum is
Ethanol NEGATIVE POSITIVE
diluted and mixed with latex particles that have
Gelation
been coated with anti-fibrin split products.
- Determines presence of split or degradation
products but will not identify whether this split
HEMATOLOGY 2

THROMBOTIC DISORDERS
THROMBOSIS: platelet or fibrin clots

PHYSIOLOGIC THROMBOSIS: normal response due to


injury.

PATHOLOGIC THROMBOSIS

- Abnormal that may lead to obstruction of


circulation.
- Can be acquired or inherited.
- Risk factors:
o Acquired conditions are related with life
events such as age, smoking and other
diseases.
o Inherited conditions are related with
congenital deficiencies in the regulatory
proteins (inhibitors) and defective
factors.

TYPES OF PATHOLOGIC THROMBOSIS

a. ARTERIAL THROMBOSIS
- Composed of many platelets with small amounts
of fibrin, RE and white cells – “white clot”.
- CAUSES: Hypertension, Hyperviscosity,
qualitative platelet abnormalities and
atherosclerosis.

b. VENOUS THROMBOSIS
- Few platelets and blood cells, and MANY FIBRIN
- Composed of large amounts of fibrin and red
cells.
- CAUSES: Abnormalities: associated with slow
blood flow (hyper viscosity of blood), activation
of coagulation system, platelet dysfunction,
leukocyte activation: produces a lot of adhesive
molecules, anatomical abnormalities of blood
vessel wall, impairment of the fibrinolytic
system, and deficiency of physiologic inhibitors
- RISK FACTORS: Life events: age, smoking, DM,
hypercholesterolemia.
HEMATOLOGY 2

INHERITED THROMBOTIC NOTES ASSOCIATED ACQUIRED


DISORDERS CONDITIONS
Anti-thrombin (AT) Deficiency Recurrent venous thrombosis DIC, liver disease, nephrotic
AT-III: main inhibitor of thrombin syndrome, oral contraceptives and
(continuous activation of V, VIII, XI, pregnancy
XIII)
Heparin Cofactor II Deficiency Not associated with thrombosis
Regulatory Proteins important in inactivation of V and VIII
Protein C Deficiency Thrombopoietin Vitamin K deficiency, liver disease,
malnutrition, DIC and warfarin
therapy
Protein S Deficiency Vitamin K deficiency, liver disease
and DIC
Activated Protein C Resistance Factor V Leiden is not inactivated  excessive clot formation
(Factor V Leiden) Mutated factor V  Resistant to inactivation of activated Protein C  Va remains
to be activated
Prothrombin Mutations Increase in the concentration of plasma prothrombin
Other Inherited Thrombotic Disorders:
 Elevated activity levels of Factor VIII are associated with venous thrombosis embolism
 Factor XII deficiency: XIIa serves as plasminogen activator  deficiency: decrease production of plasmin and
persistence of clot in the circulation
 Dysfribrinogenemia
 Hyperhomocysteinemia
 Tissue Factor Pathway Inhibitor Deficiency: TFPI main inhibitor of tissue factor pathway
THROMBOTIC THROMBOCYTOPENIC PURPURA (TTP)
 Deficiency of ADAMTS-13 (metalloproteinase enzyme important for the cleavage of ultra large vWF)
 activation of platelets  platelet plug  obstruction

HEMOLYTIC UREMIC SYNDROME


 Associated with diarrhea
 Caused by verotoxin or shigatoxin producing bacteria: E.coli 0157:H7 or Shigella strain
 This toxin will cause damage to glomerular capillaries and activation of platelets  hemolytic anemia;
fragmentation of RBC
COMMON SIGNS: TTP and HUS are fever, petechiae, neurologic signs (more often occur in TTP) and renal diseases
(more often occur in HUS); this happens when platelet aggregates is present in small circulations.
HEMATOLOGY 2

ACQUIRED THROMBOTIC DISORDERS - It inhibits coagulation by combining with


1. LUPUS ANTICOAGULANT / ANTIPHOSPHOLIPID antithrombin-III in inhibiting thrombin and Xa.
SYNDROME
- Lupus anticoagulant does not inhibit in vivo 2. ORAL ANTICOAGULANTS: COUMADIN,
coagulation but may cause prolonged in vitro WARFARIN, DICUMAROL
tests. - These inhibit vitamin K-dependent factors and
other vitamin K-dependent proteins
2. HEPARIN-INDUCED THROMBOCYTOPENIA - They alter hepatic synthesis resulting to inability
- Development of antibodies against heparin- of the liver to carboxylate the glutamyl residue
platelet factor 4 complex. of the factor leading to their functional
- This immune complex causes platelet activation, deficiency. These factors / proteins formed are
platelet microparticles, thrombocytopenia, and referred to as PIVKA/des – gamma-carboxy
hypercoagulable state. proteins.
- Coumadin (warfarin): crosses the placenta and
NOTE:
is present in human milk.
ANTICOAGULANT THERAPY
- Therapy is monitored by PT / international
Prevention of thrombosis: can be anticoagulant
normalized ratio (INR).
drugs, antiplatelet or thrombolytic drugs
- When overdosed, high levels vitamin K reverse
ANTIPLATELET DRUGS – prevent activation and the action.
aggregation and are most effective in the treatment of - For long term management.
the arterial diseases; platelets are more often
THROMBOLYTIC DRUGS
associated.
- Used to break down fibrin clots to restore
1. ASPIRIN – MOST COMMONLY USED: Inhibits vascular function and to prevent loss of tissues
Thromboxane A2 synthesis by irreversibly actetylating and organs.
the enzyme cyclooxygenase; acetylsalicylic acid; - Also used in acute arterial thrombosis for
irreversibly affects platelet function by inhibiting the
immediate thrombolysis.
cyclooxygenase (COX) enzyme and thereby the
formation of thromboxane A2 (TXA2). SERVE AS PLASMINOGEN ACTIVATORS
2. OTHER ANTIPLATELET DRUGS: dipyridamole,
thienopyridines, ticlopidine and clopidogrel. 1. UROKINASE – this is not a fibrin specific; it is used in
the treatment of venous thromboembolism, MI and
thrombolysis of clotted catheters.
ANTICOAGULANT DRUGS
2. STREPTOKINASE – not fibrin specific.
- Inhibit thrombin and fibrin formation.
- Prevention of venous thrombosis.

1. INTRAVENOUS ANTICOAGULANT: HEPARIN


- The anticoagulant activity of heparin is enhanced
by binding to AT.
- Heparin-AT complex inactivates thrombin and
Factor Xa.
- Heparin dosage is monitored by APTT &
activated clotting time.
- When overdosed, its action is reversed by
protamine sulfate.
- Effect is immediate because it enters directly
into the vessel.
HEMATOLOGY 2

RECAP: PRIMARY AND SECONDARY HEMOSTASIS

PATTERNS OF CLINICAL BLEEDING IN DISORDERS OF HEMOSTASIS


PRIMARY HEMOSTASIS SECONDARY HEMOSTASIS
Characteristics platelet/vascular problem coagulation factor problems or presence of
inhibitors and systemic disease
Onset Spontaneous bleeding, immediate after trauma Delayed after trauma
Sites Skin, mucous membranes Deep tissues
Form Petechiae, ecchymosis Hematomas (bluing of skin with tumor/swelling
 clot within tissues
Mucous Common (nasal, oral, gastrointestinal, Less common
membrane genitourinary)
Other sites Rare in deep tissues Joint, muscle, central nervous system,
retroperitoneal spaces
Clinical examples Quantitative Platelet Defects: Factor deficiency, liver disease, acquired
 Thrombocytopenia inhibitors
Qualitative Platelet Defects: (adhesion,
aggregation or release reaction, vascular defects)
 platelet defects, vWD, scurvy

DISORDER OF COAGULATION, FIBRINOLYSIS AND THROMBOSIS


INHERITED DISORDERS OF COAGULATION
COAGULATION FACTORS DEFICIENCY

All CF are produced by the liver except PF3, CF IV(Ca++) and vWF
FACTOR INHERITED COAGULOPATHIES ACQUIRED NOTES
INHERITANCE COAGULOPATHY COAGULOPATHY
PATTERN
PROTHROMBIN GROUP
II Autosomal Prothrombin Liver disease Least to occur
recessive deficiency Vitamin K deficiency
VII Autosomal F VII deficiency (II,VII,IX,X)  functional Most profound
recessive deficiency; no gamma disorder/deficiency;
carboxylation shortest circulating life 
first to disappear.
Oral anticoagulant PT: immediately prolonged;
therapy: inhibits vit. K  more sensitive to liver
functional deficiency diseases and vit. K.
X Autosomal F X deficiency Bile duct obstruction (gall
recessive stone)  incomplete
IX Sex -linked digestion and absorption Hemophilia B; Christmas
of vit. K  functional Disease
deficiency
FIBRINOGEN GROUP
I Autosomal Afibrinogenemia Severe liver disease Rarest to occur
recessive DIC
HEMATOLOGY 2

Autosomal Dysfibrinogenemia Fibrinolysis Consumable factors during


dominant Normal levels of I but clotting
has abnormal Sensitive to high levels of
structure plasmin
V Autosomal Owren’s disease
recessive Labile Factor
Deficiency
Parahemophilia
XIII Autosomal F XIII deficiency
recessive
VIII Sex linked Inflammatory liver
(hepatic) diseases
 factor VIII increases
CONTACT GROUP
XI Autosomal Most common among
recessive contact factors: Hemophilia
C but least likely to occur in
hemophilia
XII Autosomal F XII deficiency; 2nd most common; not
recessive Hageman deficiency related to bleeding but
related to
thrombosis/abnormal clot
formation
PK Autosomal Fletcher trait
recessive
HK Autosomal Fitzgerald trait
recessive

HEMOPHILIAS
HEMOPHILIA

- Love of hemorrhage
- Inherited disorders characterized by a deficiency of the anti-hemophilic factors.

HEMOPHILIA A B C
FACTOR DEFICIENT VIII (VIII:C) IX XI
OTHER NAME Royal disease and Classic Christmas disease Rosenthal syndrome
hemophilia
INHERITANCE Sex-linked Sex-linked Autosomal
OCCURENCE 1 in 5,000-10,000 males 1 in 25,000 -30,000 in males Ashkenazi Jews
REGARDED AS Most severe congenital 2nd most severe congenital Least and rarest to occur
bleeding anomaly bleeding anomaly same S/s
with A but milder
PROLONGED TEST APTT APTT APTT
TREATMENT Commercial Factor VIII Factor IX concentrate Fresh frozen plasma (FFP) or
concentrate (prevent (prevent bleeding) whole blood: Replenish
Does not correct bleeding) Recombinant F-IX ( in deficient factor
hemophilias but only Recombinant F-VIII (in developed countries: No single blood component
ARREST BLEEDING or developed countries: standard standard treatment and
treatment and management) management)
HEMATOLOGY 2

PREVENT POSSIBLE DDAVP (1-desamino-B-


BLEEDING arginine-vasopressin)

Contraindication: ASPIRIN,
ANALGESICS (anti-platelets)

Notes: Royal disease: first observed in Named after Stephen Rosenthal: means “rose-
royal family of England (Queen Christmas; case published valley”: German compound
Victoria: Carrier) (December 22, 2011) in The
New England Journal of
Medicine

1. HEMOPHILIA A/CLASSIC HEMOPHILIA with major trauma or


- A sex-linked disorder characterized by a surgery
deficiency of Factor VIII:C
- Most common hemophilia
- Clinical signs and symptoms depend on NOTE:
remaining factor VIII:C level  Generally applies to both Factor VIII and factor
IX deficiencies; may not apply to deficiency of
- Therapy:
other factors.
o Commercial Factor VIII concentrate
 To determine the remaining factor level 
o DDAVP (1-desamino-B-arginine-
Factor Assay (+ Factor VIII –deficient plasma
vasopressin) or + Factor IX-deficient plasma)
 Bleeding manifestation: Hematoma,
2. HEMOPHILIA B (CHRISTMAS DISEASE) hemarthrosis  severe hemophilias
- Sex-linked disorder characterized by a deficiency
of Factor IX
- Second most common hemophilia with clinical
signs and symptoms similar to hemophilia A HOW IS SEX-LINKED HEMOPHILIA (A AND B)
- Therapy: Factor IX concentrate TRANSMITTED?
- Applies PUNNET SQUARE
SEVERITY OF HEMOPHILA A AND B - 50:50 Rule of inheritance of genes including
the sex chromosomes( XX:Female; XY: Male)
- Severity of bleeding depends on factor level For females:
which classify hemophilias as severe, moderate, XXH: Only one chromosome is affected  CARRIER
or mild; applicable only to sex-linked hemophilia:  Carrier (does not manifest s/s of hemophilias
A and B but can transmit abnormal gene to either son
or daughter)
CLASSIFICATION FACTOR COMMON XHXH: When both X chromosome have the abnormal
OF BLEEDING LEVEL MANIFESTATION gene  HEMOPHILIAC
Severe <1% Spontaneous bleeding;  Happens only if father is hemophiliac and
bleeding with minor mother is either hemophiliac or carrier
surgery or trauma XHO: One X chromosome is inactivated; Female is
Moderate 1–5% Spontaneous bleeding suffering from Turner’s syndrome  HEMOPHILIAC
uncommon; may bleed
with surgery or trauma For males:
Mild 5- 20% No spontaneous XHY: Once X is affected  HEMOPHILIAC (one’s that
bleeding; may bleed have the disease and shows s/s)
HEMATOLOGY 2

 HEMOPHILIAC (one’s that have the disease  Swelling and hematomas after blood
and shows s/s) extraction or IM injection
 Muscle tightness

3. HEMOPHILIA C (ROSENTHAL SYNDROME)


- An autosomal recessive disorder characterized
by a deficiency of Factor XI
- Has an increased incidence among Ashkenazi
Jews
- Bleeding is mild unless stressed by trauma or
IMAGE 1: Union of Carrier Mother and Normal Father surgery
will produce sons: (50% normal and 50% - Considered as silent type
haemophiliac) and daughters: (50% normal and 50% - Not associated with bleeding disorder or just
carrier) mild bleeding disorder: explain by current
IMAGE 2: Union of Normal Mother and Hemophiliac coagulation pathway
Father will produce sons: (100% normal) and - Therapy: Fresh frozen plasma or whole blood
daughters: (100% carriers)
4. PREKALLIKREIN ; HMWK ; FACTOR XII
DEFICIENCY
- Autosomal recessive disorders with no
associated bleeding tendencies.
- Patients are more vulnerable to excessive
clotting (thrombosis).

ACQUIRED DISORDERS OF COAGULATION


- No genetic abnormality or anomaly has been
associated with these condition. Gene is
something that underwent mutation due to
some mutagens. Main causes are the other
underlying conditions or diseases.

NOTE: 1. INHIBITORS
HOW TO DETECT HEMOPHILIA A. NON-SPECIFIC FACTOR INHIBITORS
 Signs and symptoms - Directed not against coagulation factor but
 Presence of FAMILY HISTORY of bleeding other molecules and proteins involved in
among male relatives in the mother’s side coagulation.
 Laboratory tests: APTT and Clotting Factor - Example: Lupus Anticoagulant (LA)
Assays
NOTE:
EARLY CLUES FOR THE DETECTION OF HEMOPHILIA LUPUS ANTICOAGULANT (LA)/ ANTIPHOSPHOLIPID
 Prolonged bleeding after cutting the umbilical ANTIBODIES (APA)/ ANTICARDIOLIPIN ANTIBODIES
cord  A non-specific inhibitor directed against
 Hematomas and joint swelling in babies platelet phospholipid and phospholipid-
beginning to creep, crawl and walk protein complexes.
 Limited range of motion of certain joint  Its presence will prolong
 Refusal to use limb (young child)  Directed to epitopes of proteins bound to
 Prolonged bleeding after circumcision phospholipids  THROMBOSIS
 Prolonged bleeding after tooth extraction
HEMATOLOGY 2

 Prolonged: APTT (effect is less common) and  Decrease intake of vit. K.: Malnutrition;
dilute Rusell Viper Venom Test (dRVVT) source of Vit. K is diet (green leafy
o The combination of these two tests vegetables)
serves as a screening test for the  Use of oral anticoagulant (warfarin,
presence of Lupus Anticoagulant. coumarin, dicoumarol and other
 Immuno assays (ELISA): for confirmatory test derivatives) or presence of Vitamin K
as well as for titer.
antagonist: inhibits vitamin K 
 Not at risk from suffering bleeding conditions.
inhibiting normal synthesis of vit. K
Instead, they are more at risk to suffer from
dependent coagulation factors 
THROMBOSIS.
referred as PROTEINS INDUCED BY
VITAMIN K ANTAGONIST (PIVKA) 
B. SPECIFIC FACTOR INHIBITORS abnormal in function
o These are IgG autoantibodies  Decreased normal flora in the GIT
directed against specific (bacteroides fragilis, E. coli); prolonged
coagulation factors (e.g. Factor use of strong antibiotics: broad
VIII:C, IX, X and Factor XI inhibitors) spectrum antibiotics  damage normal
flora  growth of abnormal flora
NOTE:
 Decreased intestinal absorption intake
FACTOR VIII:C INHIBITOR
 Most likely to occur  Intestinal infection and Biliary duct
 Commonly developed from patients suffering obstruction (obstructive jaundice).
from B-cell malignancies, autoimmune  Vitamin K is fat soluble vitamin it
diseases (Systemic Lupus Erythematosus), and requires bile for its digestion thus
elderly. obstruction along the normal flow of bile
 Production may also be induced in patients which can be caused by the presence of
who have received the Factor VIII tumor or gallstone that constricts this
concentrate. bile duct and cause obstruction to bile
 Present bleeding conditions similar with flow  inadequate digestion and
Hemophilia A (Acquired hemophilia). absorption.
 Prolonged APTT and will serve as a screening
test to detect VIII:C Inhibitor. NOTE:
 Bethesda Assay-titer: determine the titer of  Infants: given vitamin K at birth to
antibody. improve the baby’s synthesis of vitamin
 High-dose Factor VIII: given to prevent from K dependent factors; intestine is sterile.
suffering bleeding conditions. Bacteria (from maternal skin) grows
 Immunosuppressant drugs (Cytoxan, only when the baby start to breastfeed.
Rituximab): given for longer management of  Importance of Vitamin K: gamma
the patient. It is used to inhibit or prevent the carboxylation of II, VII, IX, X; the addition
immune system from continuous production of this carboxyl radicals to their terminal
of antibody against factor VIII. glutamic acid will cause them to gain a
negative charge thus will allow them to
interact with Calcium which is positively
2. VITAMIN K DEFICIENCY charged and on bind to platelet
- More on functional deficiency and less on phospholipid which is negatively
synthesis deficiency charged.
- Will result to functional decrease of Factor II, VII,
IX, and X, protein C, S and Z
- Causes of Vitamin K Deficiency:
HEMATOLOGY 2

3. LIVER DISEASE - CAN OCCUR IN ANYONE, NO AGE GROUP NOR


- Both clearance and synthesis function will be GENDER PREFERENCE.
affected  Bleeding disorders. - Occurs as complication of another disorder 
- Leads to synthesis deficiencies (FVII deficiency is causes release of thromboplastic substance 
the most profound; first to decrease due to its initiates both Coagulation and Fibrinolysis: both
short circulating lifespan) system simultaneously occurs and may either
- Decrease clearance activity of activated predominate the other.
components including plasminogen activators
- Increase in FSP due to increase fibrinolysis
- DECREASES: VII (first), PK, I
 Factor VII: most profound to decrease.
When the liver is unable to produce
coagulation factors, Factor VII is the first
to decrease due to its short circulating
life span.
 PK: one of the first to decrease
 Factor I (Fibrinogen): last to decrease
- INCREASE: VIII; if the liver disease is  Result from the liberation of thromboplastic
inflammatory in nature, Factor VIII increases. substance that activates coagulation
 Release of thromboplastic-like substance may
NOTE: be secondary to trauma, sepsis or toxin that
Liver functions in the synthesis of all coagulation
tissues suffering from this may cause the release
factors except calcium and tissue factor and in
of inflammatory substances that may then exerts
clearance, it removes activated coagulation
thromboplastic-like activity
components to prevent further activation and
excessive coagulation. It also removes  Complication of childbirth obstetrics as
plasminogen activators to prevent excessive placental tissue are separated, they release
fibrinolysis or excessive production of plasmin. tissue fluids that can exert thromboplastin-like
It removes products of fibrinolysis: Fibrin split activity, so the substance will activate both
products. coagulation and fibrinolysis.
 Plasminogen is also activated through the
contact phase (TPa)
4. MASSIVE TRANSFUSION
- Replacement of more than 1.5 L blood volume NOTE:
or more than 3 units of blood bags in 24 hours Thus, coagulation and fibrinolysis occur
resulting to dilution of coagulation factor; simultaneously and either may dominate, resulting to:
DILUTIONAL COAGULOPATHY (in transfusion of  Consumption of coagulation factors (I,V, VIII
pRBC and NSS) and increased introduction of and XIII) and platelets as thrombi are formed
anticoagulants (each unit contains 63 mL of and deposited locally and widely in the
anticoagulant; 10% or more enough to interfere circulation
to normal coagulation). o As COAGULATION is activated,
platelet aggregates  they are
consumed resulting to
5. DISSEMINATED INTRAVASCULAR
thrombocytopenia and bleeding.
COAGULATION (DIC) The platelet aggregate will also
- Disseminated: clot formation occurs elsewhere obstruct normal circulation 
in the body thrombosis  vasoocclusion
- Often described as CONSUMPTION because of thrombus within the
COAGULOPATHY circulation. Because of the clot
deposited in the tissues, the
HEMATOLOGY 2

normal flow of oxygen and blood Hematom II, VIII, IX Umbilical X, XIII
will be obstructed resulting to as cord
ischemia or decrease in tissue bleeding
oxygenation  tissues die Mucosal II, VIII, IX, XI Miscarriag I, XIII
(necrosis): kidneys, lungs, liver bleeding e
and brain. Hemarthr VII, IX, X Thrombosi Abnormal
 RBC fragmentation  SCHISTOCYTES osis s fibrinogens;
o Clot deposits cause trauma to red Postsurgic I, II, V, VII, LA;
cells; some are caught or slice/cut al VIII, IX, X, inhibitor
off by fibrin strands  bleeding XI, XIII deficiencies
fragmentation or lysis  Intracrani VII, VIII, IX, Asymptom FXII, PK, HK
hemolysis lead to presence of RBC al XIII atic
fragments called schistocytes on bleeding -They do
circulation and blood smear. not play a
 Increased in FDP and D-dimer major role
o FIBRINOLYSIS ACTIVATION: in in vivo
production of fibrin coagulatio
split/degradation products: XYDE n
 interferes in normal platelet NOTE:
function as well as in the Factors VIII and IX is the most severe coagulation
polymerization of fibrin factor deficiency.
monomers  affects normal
coagulation  more bleeding will
occur. CYTOCHEMICAL STAINS
CYTOCHEMISTRY

NOTE: DIC - Microscopic study and identification of chemical


LAB FINDINGS: all laboratory test will appear constituents within individual cells. It is useful in
abnormal like platelet count (decrease: the identification of malignant cell types on the
thrombocytopenia) and prolonged bleeding time and basis of cytoplasmic or nuclear chemistry;
clotting time(PT, APTT, TT, ST) cellular constituents that are present in
abnormal form or amount; lack of cellular
TREATMENT: underlying cause of DIC should be constituent; and cells exhibiting functional
considered abnormalities.
 Plasma and platelet transfusions – restore
consumed BRIEF INTRODUCTION TO LEUKEMIA
 Heparin –prevent further activation of
clotting mechanism - Leukemia refers to group of disorders
- In the category of cell line, leukemia can be
differentiated to either Myeloid or Lymphoid
Leukemia
CLINICAL MANIFESTATIONS OF COAGULATION - In the basis of Blast cell
FACTOR DEFICIENCIES o Acute Lymphocytic Leukemia: Pre-
dominance of lymphoblast.
TYPE OF COAGULATI TYPE OF COAGULATI o Chronic Lymphocytic Leukemia:
BLEEDING ON FACTOR BLEEDING ON FACTOR Pre-dominance of mature with few
DEFICIENCY DEFICIENCY lymphoblast.
Easy II, VIII, IX Delayed I, XIII o Acute leukemia: refers to a type of
bruising wound leukemia with poorer prognosis with
healing
HEMATOLOGY 2

an increase predominance of blast acid except the isoenzyme number 5


cells or immature cells which is the one present in hairy cell
o Chronic Leukemia: the decrease of leukemia.
blast cells and increase of mature - In Chronic Myeloid Leukemia the predominant
cells. cells are the mature myeloid cells especially the
o Predominance of myeloblast is granulocytes. CML is also called Chronic
associated with Acute Myeloid Granulocytic Cells. Granulocytes can range from
Leukemia 50,00-300,000
o Predominance of mature myelocyte - Having a very high WBC count can also be
is associated with Chronic Myeloid observed in Leukemoid Reaction which is due to
Leukemia underlying condition.
- Blast cells are not easily identified because of - Both leukemoid reaction and chronic myeloid
their common characteristics (larger in size, large leukemia are characterized by a very high WBC
nucleus, smaller and deeply basophilic count. However, Leukemoid Reaction is
cytoplasm), cytochemical stains are used to reversible that when the underlying condition is
differentiate in the absence of CD marker resolved, the WBC will return to normal count.
- Acute Myeloid Leukemia can be further CLM is a permanent condition. To differentiate
classified knowing it is a stem cell. Leukemoid reaction from CML, alkaline
- French American British System classified phosphatase is used.
leukemia based on the morphology of the cells
ENZYMATIC STAINS
after staining with Romanowski stain and their
PEROXIDASE/MYELOPEROXIDASE (MPO)
reaction to cytochemical stain.
- FAB further classified ALL as ALL1, ALL2, ALL3. - The stain itself does not contain the peroxidase
While AML is classified into seven categories enzyme. The peroxidase enzyme is inside the
(AML1-AML7). cell, the composition of the stain is the substrate
- In AML for peroxidase enzyme
o AML1-AML3 predominance cell is - Myeloperoxidase is a normal constituent of the
Myeloblast, promyeolocyte myelocyte primary granules of the myelocytic cells which is
(granulocytic series) observed as early as promyeolocyte.
o AML4 and AML5 uses alpha naphthyl - Stain marker for primary granules & auer rods
Chloroacetate esterase (fused primary granules)
o AML4 stain positive in both Specific and - Stain positive in AML but negative in ALL
non- specific esterase because it - Differentiates AML from ALL
contains both granulocytic and - Stain marker for immature myeloid cells
monocytic cells - Stain positive in granulocyte but not in
o AML5- is the acute monocytic leukemia lymphocytes
o AML6 is called Erythroid Leukemia, - Positive Activity: reddish-brown deposits (in
leukemic cells are precursors of cytoplasm of granulocytes and monocytes)
erythrocyte. Positive in Periodic Acid - Note: Myeloperoxidase enzyme deteriorates;
Schiff. Stain should be done only on fresh specimens
o AML7 leukemia of megakaryocytes
called acute megakaryoblastic leukemia. ALPHA-NAPHTHYL ACETATE ESTERASE & ALPHA-
- One example for Chronic Lymphocytic Leukemia NAPHTHYL BUTYRATE ESTERASE (NON-SPECIFIC
is the Hairy Cell Leukemia which affects B-cells. ESTERASES/NSE)
Hairy cells contain ACP. - Used to detect the granulocyte of monocytic
o ACP isoenzyme is present in all blood origin
cells and can be inhibited by tartaric - Nonspecific because it can be seen in other cells
HEMATOLOGY 2

- The stain does not contain esterase but rather NAPHTHOL ASD CHLOROACETATE ESTERASE (SPECIFIC
substrate for the enzyme ESTERASE)
- If the cell contains the enzyme it will catalyze the
 Marker for mature & immature Neutrophil and
hydrolysis of the butyrate or acetate in the stain
mast cells.
- Unfixed sample can be used so long in the dark
for as long as 2 weeks  Substrate is the chloroacetate
- Marker for cells of Monocytic origin  Use to identify immature or primitive
- Other cells (+): Megakaryocytes, Platelets, granulocyte ALM1-ALM3
Histiocytes, Plasmacytes, some T-lymphocytes  Positive Activity: bright red granules in
- Positive Activity: red-brown/dark red cytoplasm.
 This enzyme is stable and may last for months.
LEUKOCYTE/NEUTROPHIL ALKALINE PHOSPHATASE
(LAP/NAP) TERMINAL DEOXYRIBONUCLEOTIDYL TRANSFERASE
(TDT)
- Stains NEUTROPHILS (the only leukocytes that
contain this activity)  Stains DNA polymerase immunoperoxidase
- Neutrophils contain various amount of ALP  Marker for immature Lymphoid cell
- Principle: Differential count involving  Positive in almost 90% cases of ALL
neutrophils  Also useful in the detection of the
- Differentiates Leukemoid reaction (LR) from “lymphoblastic transformation” of chronic
Chronic myeloid leukemia (CML) myeloid leukemia (CML)
- Reference Value: 30 –185 LAP score  Detects blastic transformation of CML
- Increase LAP score is observed in the following: o CML can transform into an acute
o during the last trimester of pregnancy - leukemia
Hodgkin disease o Acute leukemia more than or equal to
o Polycythemia vera - Multiple myeloma 30% blast cells
o Aplastic anemia - Obstructive jaundice o CML can transform either AML or ALL.
- To compute, multiply the counted neutrophils by  AML: (+) MPO stain and SBB
their grade.  ALL: (+) TdT, it means that it
- Normal to High: Leukemoid reaction transforms into ALL.
- Below than normal value to zero: CML
ACID PHOSPHATASE
TARTARIC ACID RESISTANT ACID PHOSPHATASE (TRAP)
 Present in all hematopoietic cells and found in
- For the diagnosis of Hairy cell leukemia (HCL) lysosomes
- General Principle: ACP is Detected in almost all  Activity is indicated by purple to red granules
blood cells but when treated with tartaric acid it  Unstable, cannot be stored.
is inhibited except isoenzyme number 5
- Labile if unpreserved NON-ENZYMATIC STAINS
- Sample should first be fixed and be stored at - PRUSSIAN BLUE STAIN
20°C and can be used atleast 2 weeks  Stains siderotic granules (for the diagnosis of
- Other Tartrate Resistant cells: Sezary cells; sideroblastic anemia).
Histiocyte; T-cell of acute lymphocytic leukemia  Used to detect hemosiderin in urine
- Activity is indicated by purple to dark red  Used to identify iron in the ferric state (Fe+3)
granules in cytoplasm

CYANIDE-RESISTANT PEROXIDASE

- For identification of Eosinophilic components


- Positive Activity: brown
HEMATOLOGY 2

PERIODIC ACID SCHIFF (PAS) TOLUIDINE BLUE O

 Stain reacts w/ aldehyde groups in glycogen,  A metachromatic stain


mucoprotein, glycoproteins, and high molecular  Binds with mucopolysaccharides in blood cells
weight carbohydrates.  For the recognition of basophils and mast cells
 Does not detect stains instead it combines with  (+) Result: Reddish-Violet
the aldehyde group of glycogen.
 Positive to all blood cells except normal ADDITIONAL NOTES:
Primary fibrinolysis/fibrinogenolysis: Increase
erythroblast or pronormoblast
activators of plasminogen  increased plasmin: not
 Positive erythroblast indicates that it is affected
normally present
by AML6. Secondary fibrinolysis: Release of thromboplastin-like
 Positive in erythroblasts in Di Guglielmo disease substance  activates both coagulation and
/ Erythroleukemia. fibrinolysis  increased plasmin.
 L1 and L2 also stain positive in PAS
 The positivity of L1 to Pas is described to be Naturally occurring inhibitors: naturally present in the
chunky or block-like positivity circulation and their purpose is for normal regulation
 L2 is not that large as L1 of coagulation mechanism. They maintain
homeostasis. Examples: AT-III; TFPI, Protein C, Protein
 Negative Activity: bright fuchsia pink (Pattern of
S, alpha-2 macroglobulin, alpha-2 anti-plasmin, etc.
staining varies with each cell type)
 Also differentiates Acute Lymphoblastic Abnormally, we could also develop inhibitors or
Leukemia. autoantibodies. Under ACQUIRED COAGULATION
 Peroxidase of eosinophil is resistance to cyanide DISORDER; not normally present in the system or
 Cyanide resistance peroxidase is used to identify circulation but we produce them when our system is
peroxidase of eosinophils abnormally activated.

INHIBITORS: autoantibodies that cause prolongation


SUDAN BLACK B (SBB) of certain test because they interfere in normal
coagulation.
 Marker for phospholipids and lipids
 Has the same staining reaction as 2 CATEGORIES:
myeloperoxidase 1. SPECIFIC
 Gives the same information as Peroxidase in the  inhibitor directed against specific coagulation
interpretation. factor like FVIII, FIX or FVII inhibitor FACTOR
 Positive in AML. VIII: C INHIBITOR
 Advantage over myeloperoxidase is that it is  Most common and severe coagulation factor
more stable, because myeloperoxidase detects inhibitor in VIII:C portion not the complex
enzyme which is somewhat labile and may  Inhibit Factor of VIII: C only: does not have an
disappear in prolonged storage. Sample stained effect on vWF.
in SBB can give reliable result for months.  Prevents VIII:C in participating as cofactor in
 Differentiates AML from ALL the formation of intrinsic tenase.
 Normally present in elderly with B-cell
 (+) Result: Dark purple-black granules
anomalies
NOTE:  Patients are high risk for suffering bleeding
 Can be done on stored specimens.  Prolonged: APTT
 More sensitive than chloroacetate in the  Bethesda assay – titer: antibody
demonstration of mature & immature measurement
neutrophils and mast cells.  Acute management  high-dose F VIII  not
all will be inhibit  coagulation proceeds (for
short term only).
HEMATOLOGY 2

 Long term management  MYELODYSPLASTIC, MYELOPROLIFERATIVE and


Immunosuppression  patient is given LYMPHOPROLIFERATIVE DISEASES
cytosan  inhibit production of all antibodies: MYELODYSPLASTIC SYNDROME
should be well-planned.
 myelo – bone marrow; dys – abnormality; plastic
2. NON-SPECIFIC – development  abnormality in development
 directed against factor or protein involved in of cells arising from bone marrow.
coagulation but is not specific to a certain
MYELOPROLIFERATIVE DISEASE
coagulation factors; targets anything.
LUPUS ANTICOAGULANT/APA/ ANTICARDIOLIPIN  Abnormal proliferation of cells in the bone
ANTIBODY marrow.
 Inhibits phospholipids and proteins bound to
phospholipids but it has no anticoagulant MYELODYSPLASTIC-MYELOPROLIFERATIVE
effect in vivo  does not lead to bleeding.
 Directed to epitopes of proteins bound to  Abnormal proliferation and development of cells
phospholipids  Thrombosis; abnormal in the bone marrow.
clotting NOTE:
 Seen in immunocompromised patients: NORMAL LIFE: cell production = cell destruction 
higher in patients suffering from SLE, HIV, and NORMAL cell count
other autoimmune disorders that affects B- Main parent cell: PPSC  Stem cell: LSC (T and B
cells production lymphocytes and non-A non-B population) and MSC
 Effect is different in vivo and in vitro (RBCs, Platelets, Monocytes and granulocytes)
 In vitro: has anticoagulant effect and causes UNILINEAGE DYSPLASIA: only one cell line is affected
prolongation of clotting time tests like APTT MULTILINEAGE DYSPLASIA: one or more cell line is
and dRVVT; less likely in PT affected
 Immunoassay: for diagnosis  ELISA
HEMATOLOGY 2

MYELODYSPLASTIC MYELOPROLIFERATIVE MYELODYSPLASTIC-


Myelodysplastic Syndrome (MDS) MYELOPROLIFERATIVE
or Dysmyelopoetic Syndrome
(DMS)
abnormality in development of abnormal proliferation of cells in the bone abnormal proliferation and
bone marrow cells marrow development of cells in the bone
marrow
MAIN ABNORMALITY: MAIN ABNORMALITY: Uncontrolled Can’t classify as MDS or MPD alone
Hypercellular maturation  cells proliferation of one or more of the BM  MDS/MPD  with variable
are abnormally large of cells Dysplasia might be present (little) increases in cells as well as
myelo-cell line (RBCs and WBCs but not common cytopenia and morphologic
but NOT LYMPHOCYTES) dysplasia
Abnormal morphology: RBC and Abnormal number of cell: RBC and WBC Show mixed characteristics of MDS
WBC and MPD.
MAIN DIFFERENCE: Many MAIN DIFFERENCE: Little or no dysplasia
dysplasia
Apoptosis (programmed cell Proliferation outspaces apoptosis
death) predominates  easily
perish/destroyed
BLOOD PICTURE: Cytopenia  BLOOD PICTURE: Overproliferation 
decrease in blood cells increase in blood cells
Pre-leukemia DISORDERS: DISORDERS:
• Not leukemia but can 1. Leukemia: genetic involvement; 1.Chronic Myelomonocytic
progress to leukemia  complicated classification Leukemia (CMML)
px are at risk of suffering A. ACUTE LEUKEMIA 2. Atypical Chronic Myeloid
leukemia (AML) B. CHRONIC LEUKEMIA Leukemia (aCML)
• Common in elderly 3. Juvenile Myelo-Monocytic
2. Chronic Myeloproliferative Disorders
patient: Age 50 and above Leukemia (JMML)
• Acquired condition 4. MDS/MPD-U: unclassified
CLINICAL COURSE/PROGNOSIS:
survival
Shorter than CMPD longer than
Acute Leukemia  AML: short
clinical course
DISORDERS: Refractory 
unresponsiveness
1. Refractory Cytopenia Unilineage
Dysplasia (RCUD)
Refractory Anemia (RA): only red
cells are affected
Refractory Neutropenia (RN): only
neutrophils are affected
Refractory Thrombocytopenia
(RT): only thrombocytes or
platelets are affected
2. Refractory Anemia with Ringed
Sideroblast (RARS): immature
RBCs with siderotic granules
HEMATOLOGY 2

3. Refractory Anemia with


Multilineage
Dysplasia (RAMD)
Refractory Anemia with Excess
blast (RAEB)
4. 5q- SYNDROME OR
ISOLATED 5q DELETION
5. MDS-U: unclassified

TWO SYSTEMS EMPLOYED IN CLASSIFICATION OF LEUKEMIAS AND MYELOID DISORDERS

The FAB Classification The WHO Classification


French-American British System World Health Organization
Group of scientist and doctors
Basis: Morphologic appearance  Basis includes more criteria: morphology,
examination of blood smear stained with chromosomal study (genetic abnormality) and
Wright’s or Giemsa; cellular level  not immunologic probe (CD markers); more
accurate and precise genetic level of disease
FIRST BASIS CURRENT BASIS
RA Refractory anemia Refractory anemia RCUD
RARS Refractory anemia with ringed sideroblasts Refractory anemia with ringed sideroblasts
RAEB Refractory anemia with excess blasts Refractory cytopenia with multilineage dysplasia
CMML Chronic Myelomonocytic Leukemia
RAEBIT Refractory anemia with excess blasts in Refractory anemia with excess blast
transformation Type I
Type II: characteristics are similar with RAEBIT
Myelodysplastic syndrome unclassifiable
5(q) chromosome abnormality

MYELODYSPLASTIC SYNDROME (MDS)


 MDS are group of disorders that result from clonal abnormalities of hematopoietic pluripotential stem cells. These
are characterized by hypercellular maturation of the erythroid cells, granulocytes and megakaryocytes.
 They are often described as “Pre-Leukemias” FEATURES
 Occur primarily in persons over age 50
 Characterized by peripheral Cytopenias
 Also characterized by presence of dysplasia of the myeloid cell lines
 Clinical course is shorter than CMPD and longer than acute leukemias.
HEMATOLOGY 2

MAJOR CLASSIFICATION OF MDS/DMS (WHO Classification, 2008)

MDS PERIPHERAL BLOOD FINDINGS BONE MARROW NOTES


FINDINGS
Refractory Cytopenia Unicytopenia Unilineage dysplasia: Refractory:
with Unilineage Dysplasia No or rare blasts (<1% blast) only one cell is affected; Unresponsive to either
(RCUD) >10% of cells in one treatment or stimulation.
• Refractory anemia (RA): myeloid lineage Dysplastic: should be
RBCs (most common) more than or equal to
• Refractory neutropenia 10% of cells.
(RN) Main abnormality:
•Refractory hypercellular maturation
thrombocytopenia: Prognosis: >5 years; good
Platelets prognosis Risk of
acquiring
leukemia  AML (6%) 
Refractory Anemia Dyserythropoiesis: BM: <5% blasts AML: poor prognosis
megaloblastoid maturation (RA): none or <15%
(ovalocytes); ringed sideroblasts
macroovalocytes ( more than 8
micra)
PB: <1% blasts; reticulocytopenia
 increased apoptosis

Refractory anemia with Features are similar with RA. •Erythroid dysplasia only Reasons for the presence
ring sideroblasts (RARS) Dyserythropoiesis: •15% or more ringed of ringed sideroblast:
megaloblastoid maturation sideroblasts (ONLY Abnormal development
(ovalocytes); DIFFERENCE with RA) of cells and impaired RBC
macroovalocytes ( more than 8 •<5% blasts hemoglobinization: Iron
micra) ppt  siderotic granules
PB: <1% blasts; reticulocytopenia Stain to confirm presence Heme synthesis:
 increased apoptosis of iron: mitochondria; near
Anemia, no blast PRUSSIAN BLUE nucleus: iron not utilized
 siderotic granules
around nucleus forming
a ring
Prognosis: >5 years
 AML (2 years)

Refractory Cytopenia Cytopenia(s) Dysplasia in >10% of cells Multilineage: more than


With Multilineage PB: more than or equals to 10% in two or more myeloid one lineage in myelo cell
Dysplasia (RMCD) dysplasia in more than one cell lineages (neutrophil &/or line is affected; any
lines: (> 2 cell line) erythroid precursors combination
No or rare blasts (<1%) &/or Prognosis: 33 months 
<1 x 109/L monocytes; megakaryocytes <5% AML (11%)
monocytopenia is observed blasts in marrow + (less
No Auer rods than 15%) or - ringed
HEMATOLOGY 2

sideroblasts; No Auer
rods

Refractory Anemia With Cytopenia(s) Unilineage or PROGNOSIS: less than


Excess Blasts 1 (RAEB-1) <5% blasts multilineage dysplasia 2 years Incidence to
<1 x 109/L monocytes Hypercellularity progressing as AML
NO AUER RODS dyspoiesis: 5-9% blasts (25%)
without Auer rods

Refractory Anemia With Cytopenia(s) Unilineage or Incidence to progressing


Excess Blasts 2 (RAEB-2) 5-19% blasts multilineage dysplasia as AML (33%)
<1 x 109/L monocytes Hypercellularity
+ or – ; with or without: Auer rods dyspoiesis: 10-19% blasts
(fused primary granules) with Auer rods

Myelodysplastic Cytopenia(s) •Unequivocal dysplasia


Syndrome Unclassified <1% blasts in <10% of cells in one or
(MDS-U) more myeloid cell lines
PB: Rare blast when accompanied by a
cytogenic abnormality
considered as
presumptive evidence for
a diagnosis of MDS
•<5% blasts
MDS Associated With Anemia •Normal to increased 5q: long arm of
Isolated Del (5q) Usually normal or increased megakaryocytes with chromosome; there’s
Also known as: platelet count increased hypolobulated deletion of gene 
5-q Syndrome or Isolated No or rare blasts (<1%) nuclei affecting megakaryocytes
5q Deletion •<5% blasts p-arm: short arm
•No Auer rods Prognosis is good
HEMATOLOGY 2

MYELODYSPLASTIC/MYELOPROLIFERATIVE 2. ATYPICAL CHRONIC MYELOID LEUKEMIA, BCR-


DISORDERS ABL1 NEGATIVE
These are disorders that show features of both  Characterized by leukocytosis with prominent
myelodysplastic and myeloproliferative disorders. They dysgranulopoiesis  (mature and immature
are characterized by variable increases in cells as well as granulocytes)
cytopenia and morphologic dysplasia.
3. JUVENILE MYELOMONOCYTIC LEUKEMIA
TYPES:
(JMML)
1. CHRONIC MYELOMONOCYTIC LEUKEMIA  This is a clonal disorder of predominantly
(CMML) granulocytic and monocytic lineages.
 Predominant feature is persistent monocytosis  MDS/MPD: no Ph1 chromosome
of >1 X 109/L for more than 3 months duration;  Juvenile - immature or young patients
with <20% blasts (monoblasts) and few  Previously referred to as Ph1 negative CML or
promonocytes in the peripheral blood and bone Monosomy 7.
marrow with dysplasia in one or more myeloid o Normal chromosome: 23 pairs  22
cell line. pairs autosomes and 1 pair of sex
 Cytochemical stain: (+) with alpha naphthyl chromosomes; but in monosomy 7, no
acetate and alpha naphthyl butyrate esterase pair.
stains  nonspecific esterase stains  Ph : Philadelphia chromosomes; abnormal
1

 Used to be under MDS in FAB classification. translocated chromosome seen in patient


 In WHO, it is under MDS-MPD  used currently suffering CML ( leukemia and stem cell disorder
 Chronic means long/persistent or prolonged; wherein most patients but not all have Ph1 )
more than 3 months o Ph1 (+): mostly adults; when Ph1 is
present, it will fall under CML (MPD).
NOTE:
o Ph1 (-): infants or newborns and young
TYPES:
children.
 CMLL-1: <5% blasts and promonocytes in the
PB, <10% in the BM.
 CMML-2: 5-19% blasts and promonocytes in
the PB, or 10-19% in the BM  more severe
o Whenever the blast is increased, the
disease is more severe and the
poorer the prognosis

ACUTE LEUKEMIA: poor prognosis; ↑ blasts 4. MYELODYSPLASTIC/MYELOPROLIFERATIVE


CHRONIC LEUKEMIA: good prognosis; ↓ blasts
DISEASE, UNCLASSIFIABLE
ESTERASE STAINS:
 These include those cases that meet the criteria
 SPECIFIC: for immature granulocytes like
for MDS/MPN but do not fit into one of the
myeloblast  alpha naphthyl chloroacetate
esterase (combined substrate) specified subcategories.
 NON-SPECIFIC: for monocytic cells  alpha
naphthyl acetate and alpha naphthyl
butyrate esterase stains
HEMATOLOGY 2

MYELOPROLIFERATIVE DISORDERS (MPD) ENVIRONMENTAL:


LEUKEMIAS
3. CHEMICAL AGENTS: carcinogen or leukomogens
 Group of disorders: Leukemia is a disease of  Through inhalation, ingestion or simply exposure
the blood-forming tissues, predominantly the 4. IONIZING RADIATION
bone marrow. It is a malignant neoplasm  Strong dosage
characterized by disorderly, purposeless, and 5. VIRUSES: EBV and HTLV (I and II Retrovirus)
uncontrolled proliferation of one or more of the  Because of their nature (viral replication)
hematopoietic cells in the bone marrow.  EBV and HTLV (I and II Retrovirus): Burkitt
lymphoma  ALL –L3 (leukemic phase)
HEMATOPOIETIC CELLS
 Bacteria: can’t cause leukemia: reproduce or
 Medullary: bone marrow: disorder  leukemia divide by binary fission
(cancer of bone marrow of the blood forming  Virus: enters host cell  dissolve its capsid to
cells) release its viral genes or genetic component then
 Extra medullary: thymus, lymph nodes  combine with host’s cell gene  form a new
lymphoma gene  new viral elements  replicate inside
cell using their own and host’s gene  mutation.
ETIOLOGY: genetic mutation and/or oncogene activation

 Genetic mutation: cancerous gene  over 6. IMMUNOLOGICAL DEFECTS


proliferation  Patient’s immune system is suppressed 
 Oncogene activation: a gene when activated genetic mutations/oncogene activation  Prone
converts normal gene/cell into cancerous one  to suffer cancer: *Kaposis sarcoma: HIV 
Onco means cancer Breakdown of the immunosurveillance that
normally keeps neoplastic growths in check.
IMPLICATED/ASSOCIATED CONDITIONS: Leukemia  Especially seen in lymphocytic leukemia and
occur as a combination of genetic and environmental lymphoma
factors. 7. NEOPLASIA
GENETIC:

1. HEREDITY/ GENETIC
 Familial incidence: cancer that runs in the
family.
2. CHROMOSOMAL ABNORMALITY/CONGENITAL
FACTORS
 Ph1 chromosome (CML)
 Trisomy 21 (Down syndrome)  predisposed
to suffer AML
 Translocation of a part of Chromosome 8 -14 
or written as t (8:14)  Burkitt lymphoma (ALL-
L3)
o Leukemia and lymphoma are two
different terms but lymphoma can
progress to leukemia when the
cancerous cells in the lymph nodes
infiltrate the bone marrow  leukemic
phase.
HEMATOLOGY 2

NOTE:
CLINICAL FEATURES: Cancerous cell clone will
grow rapidly and the expense of normal
hematopoietic cells
Leukemic cells (LC) are cancer cells that grows
rapidly or replicate easily thus spread rapidly; found
in bone marrow  space is limited by hard bone 
LC grow rapidly  occupy the bone marrow at the
expense of the normal ones  overcrowd and
infiltrated and take up the nutrients fast
(competition) for normal cells  decreased normal
hematopoietic cells and increased abnormal cells or
LC.

CONSEQUENCES MANIFESTATIONS TREATMENT FOR LEUKEMIA


Decrease RBC production 1.Systemic Symptoms 1. Chemotherapy: destroy all cells
Decrease normal WBC production - Decreased RBC: including normal  monitor CBC
Decrease platelet production  Anemia: Fatigue, headaches, 2. Radiotherapy: radiation destroys
dizziness, paleness and others also abnormal cell.
seen in leukemia with anemia 3. Bone Marrow Transplant
- Decreased WBC: multitude of infection  - Graft vs. Host (GVHD)
fever, increase sweating 4.Differentiation Treatment:
- Decreased Platelets: Bleeding (more manipulation of growth or
important to address) and Clotting replication of cancerous cell into a
problems less or non-cancerous one  turn
into mature cell
2.Hypermetabolism - All Trans Retinoic Acid (ATRA)
5. Supportive Management:
Transfusion of blood for anemia and
Antibiotics for infections
HEMATOLOGY 2

NOTE: PROGNOSIS:
2 MAJOR CLASSIFICATION OF LEUKEMIA BASED ON  Acute: Days to months (poor; short life span)
CELL LINE:  Chronic: 1-2 years or more (good, longer life
1. Myeloid/ Non-Lymphoid/ Myeloblastic/ span)
Myelocytic/ Myelogenous Leukemia (AML or ANLL) 4 MAJOR TYPES OF LEUKEMIA
1. Acute Myeloid Leukemia (AML)
2. Lymphoid/ Lymphocytic/ Lymphoblastic/  increased in blast: myeloblast (>30%); stem
Lymphogenous Leukemia cell disorder; elderly and newborns
2. Chronic Myeloid Leukemia (CML)
BASED ON THE NUMBER OF BLAST CELLS:  decreased blast(<30%); increase mature cell:
 Upper: immature/blasts  increased in blast myelocytes
cells  ACUTE LEUKEMIA (AML or ALL) 3. Acute Lymphoblastic Leukemia (ALL)
 Acute: more blast (30 (FAB) or 20 (WHO) more  increased in blast: lymphoblast (>30%);
percent) common in children
 Lower: Mature cells increased  CHRONIC 4. Chronic Lymphoblastic Leukemia (CLL)
LEUKEMIA (CML or CLL)  decreased blast(<30%); increase mature cell:
 Chronic: less blast (20 (WHO) or 30 (FAB) less lymphocytes
percent); more mature cells
e.g. Lymphocytic/Myeloid cell line
Acute/Chronic  number of blast or prognosis/clinical
course
NUMBER OF BLAST (> or < 20/30):
 Acute: more
 Chronic: less
HEMATOLOGY 2

CLASSIFICATIONS OF LEUKEMIAS
No. of WBC (PB)
(15 x 109/L)
CHARACTERIZATION:

 LEUKEMIC: based on the number of WBC in


peripheral blood (PB)  if WBC count is higher
or more than 15x 109/L
o In leukemia, WBC/RBC count is high
however these cells are abnormal in
function because it produced by
abnormal bone marrow.
 ALEUKEMIC: leukemia if WBC count less than
15x 109/L
o Absence of leukemic or blast cells in the
circulation, leukemic cells are confined
ONLY in bone marrow
 SUBLEUKEMIC: leukemia if WBC count less than
15x 109/L found in the bone marrow and in the
circulation.

There are presence of blast and leukemic cells  both in bone marrow and peripheral blood

CRITERIA CLASSIFICATION; CHARACTERISTICS


I. Cell line 1. Myeloid/Myelocytic/Myelogenous Leukemia/Non-Lymphocytic Leukemia
2. Lymphoid/Lymphocytic/Lymphogenous Leukemia/Lymphoblastic
II. % of Blasts in 1. ACUTE LEUKEMIA:
the Peripheral  With increased blast cells in the bone marrow and peripheral blood
blood and  FAB critertia: >30% bone marrow blasts
bone marrow  WHO criteria: >20% bone marrow blasts
 Prognosis: Rapidly progressive, with a shorter prognosis of days to 6 month
2. CHRONIC LEUKEMIA
 Less than 10% blasts in PB
 With better and longer prognosis
3. SUB-ACUTE LEUKEMIA
 10-30% blast in the PB plus other symptoms
 Prognosis: at least 2-6 months
III. Number of 1. LEUKEMIC LEUKEMIA: WBC count is more than 15000/uL
WBCs in the 2. SUB-LEUKEMIC LEUKEMIA: WBC count is less than 15,000 uL, and
Peripheral  Presence of immature or abnormal cells in the PB
blood 3. ALEUKEMIC LEUKEMIA: WBC count is less than 15,000/Ul
 No immature nor abnormal cells in the PB
TWO SYSTEMS FOR CLASSIFYING LEUKEMIA
French-American British 1976; group of scientist
(FAB) Criteria: Blood smear with Romanowsky stain (Wright’s or Giemsa)  observe
Classification morphology (M1-M7; L1-L3) Cytochemical staining  identify cell line affected
>30% Acute Leukemia
World Health Improvements in 1997, 2001, 2008, 2014  classification is subject to changes
HEMATOLOGY 2

Organization (WHO) Criteria: Cellular morphology, cytochemical stains, immunologic probes of cell
classification markers (CD markers), cytogenetic or chromosomal abnormalities and clinical
syndrome
Current basis: Standard system of classifying leukemia >20%  Acute Leukemia

NOTE: ACUTE LEUKEMIA  Key myeloid antigens: Myeloperoxidase,


 Acute Myelocytic Leukemia (AML or ANLL) CD13, CD33, CD117, CD14/CD64
 AML is a stem cell disorder with  Signs and symptoms: Anemia, infection;
predominance of Blast Cells (>20%) in the bleeding (decrease in plts.); coagulation
blood or marrow and may resemble acute mechanism abnormalities (disturbance in CF)
infection at presentation. It affects all ages,
(AML –M3)
but increases with older age (>60 years). This
is also the most common form of acute  FAB: Classified into seven; requires 30% or
leukemia during the first few months of life. more blast in the myeloid series in the
 Stem cell disorder/leukemia  affect all cell marrow or circulation.
line that arise from it CFU- GEMM  WHO: requires 20% or more blast in the
 Ionizing Radiation, Leukomogens; myeloid series in the marrow or circulation
Congenital/Genetic factors (Down’s  Key myeloid antigens: Myeloperoxidase,
syndrome); Viruses, Neoplasia (cancer that CD13, CD33, CD117, CD14/CD64
transform into different or poorer type of
 Classification made by morphology,
cancer; transformation of one type of
malignancy into another form of malignancy) cytogenetics, flow cytometry and
cytochemistry  Recurrent
Clinical and Laboratory features of AML cytogenetic abnormalities that characterize
 Affects all ages, but increases with older age defined subtypes
 Two peaks: newborns or babies (almost
always AML) and elderly (>60 years)  In
children, ALL is more common
 May resemble acute infection at
presentation.
 Requires 20% blasts in blood or marrow for
diagnosis.

AML
(M1, M2, M3: MPO (+); SBB (+); aNCAE(+)); TdT
( -)
Myeloid M4 and M5: aNAE and aNBE (+)
M6: PAS (+)
Leukemia
CML
LAP (+)
Leukemia
ALL
MPO (-); SBB(-); Alpha NCAE( -); (TdT (+) L1-L3)
Lymphocytic
Leukemia L1 (intense) and L2: PAS (+)

CLL
HEMATOLOGY 2

French-American-British (FAB) Classification of the Acute Myeloid Leukemias (stem cell disorder)
CRITERIA: MORPHOLOGY AND REACTION TO CYTOCHEMICAL STAINS
M0 (AML with minimal differentiation/ Undifferentiated leukemia)
 Cells are not well differentiated
 Cytochemical staining: Negative with SBB and peroxidase  stains used to identify cells in myeloid series
(granulocytes).
MYELOCYTIC or GRANULOCYTIC SERIES
M1 (Acute Myeloblastic L. without Maturation) Myelocytic origin
No maturation: >30% myeloblast
Auer rods present (fused primary granules with pencil or
rod like shaped; spindle-shaped, red-purple)

Primary granules: promyelocyte


Secondary granules: myelocyte
M2 (Acute Myeloblastic L. with Maturation) Myelocyctic origin
With maturation: >30% myeloblast with >10%
granulocytic component (promye- to neutrophil) 
mature forms
Auer rods present
Similar to WHO: t (8:21)

M3 (Acute Promyelocytic Leukemia) With heavy granulation or Hypergranulation 


Also called Hypergranular Promyelocytic Leukemia abundant promyelocyte: primary granules of
granulocytes
Many Auer rods in bundles called “faggot cells”

WHO: t (15:17)

Associated with DIC: normal tissue (damage)  release


of thromboplastic substances  Secondary fibrinolysis:
ACTIVATION of:
Coagulation  Fibrinolysis promyelocyte lysis 
lysosomal contents  activate coagulation and fibrinolytic
cascade
MONOCYTIC SERIES [(+) with a-naphthyl acetate esterase & a-naphthyl butyrate e.]
M4 (Acute Myelomonocytic Leukemia) Myelo represents granulocytic cells
Also called “Naegeli” monocytic Leukemia Monocytic represents monocytic cells
>20% of PB WBCs are monocytes or monocytic
precursors the rest are mixture of the granulocytic cells
Predominance of both myelocytic and monocytic cells at
80:20 ratio WHO: inv (16)

M5 (Acute Monocytic Leukemia) >80% of BM elements are monocytic series; mono-, pro-
Also known as Schilling’s Leukemia and monocytes

WHO: t (9:11)
HEMATOLOGY 2

TYPES:
M5a: poorly differentiated monocytic Leukemia;
predominant cell is promonocyte (increased in BLASTS;
>80% are mostly monoblasts)
M5b: well differentiated; more of promonocytes and
monocytes (more MATURE)
ERYHTRPID SERIES
M6 (Pure Erythroid Leukemia) With neoplastic Myeloblasts & Erythroblasts
Cancerous cells in BM represents: >50% are erythroid cells
Other names: in all stages of maturation
Erythroleukemia
Erythremic Myelosis Cytochemical Stains (+) w/ Periodic Acid Schiff (PAS)
DiGuglielmo Disease PAS: stain for aldehyde; stains glycogen in the cells;
combined with aldehyde portion of the glycogen therefore
it is positive in ALL BLOOD CELLS except the normal
erythroblast or precursor or immature RBCs Normally,
erythroblast should be negative in PAS however in AML-
M6; erythroblast are abnormal  Positive in PAS.
MEGAKARYOCYTIC SERIES
M7 (Acute Megakaryocytic Leukemia) Predominantly Megakaryoblasts (>30%) and
micromegakaryoblast Cytochemical stain: Negative (-) w/
SBB and peroxidase

NOTE:
HEMATOLOGY 2

World Health Organization (WHO) CLASSIFICATION: AML

In order to classify the cell under AML they should be


positive with the following: Key Myeloid Antigens:
CD13, CD33, CD117, and CD14/CD64

Primitive Myeloblasts: (+) Myeloid associated antigens:


MPO (peroxidase enzyme in primary granules), CD13,
CD33, CD117

More mature myeblasts: produces enzymes  (+)


Cytochemical stains: MPO (peroxidase), SBB (lipids),
CAE (enzymes; esterase)

WHO Classification - Main Categories of AML;


current basis
1. AML with recurrent cytogenetic abnormalities
 Translocation (t) and inversion (inv)
 M3

2. AML with multilineage dysplasia


 There is a combination of cell line affected

3. AML, Therapy related: has mutagenic effects 


cause mutation
 Alkylating agents: chlorambucil,
cyclophosphamide, thiotepa, and busulfan
 Exposure to radiation
 Appears 5 years after exposure to both
alkylating agents and/or radiation: MDS  AML

4. AML, Not otherwise specific/classified (sub-


classified by morphology and
immunophenotype)
 FAB: M1-M7 (except M3) retained as sub-
classification

With stars: AML with recurrent cytogenetic


abnormalities (translocation and inversion) 
Myeloid associated Ag: MPO, CD13, CD33, CD117.
Second table: AML, Not otherwise specific/classified;
retained FAB
M3: under AML with recurrent cytogenetic abnormalities
 APL with t (15:17).
HEMATOLOGY 2

KEY FEATURES OF THE MAJOR ACUTE MYELOID LEUKEMIAS (AML)

Category Cell Morphology Cell surface markers Cytogenetics Prognosis


AML with t (8;21) >20% blasts, >10% CD 13, 33, 117, t(8;21)(q22;q22) More favorable
maturing granulocytes, CD19, CD34 AML1/ETO
Auer rods, dysplasia
AML with inv (16) Blasts with both CD 13,33,14,4,64 inv(16)(p13;q22) or More favorable
monocytic & t(16;16)(p13;q22)
neutrophilic
differentiation,
increased
eosinophils/immature
eosinophils
APL with t (15;17) Promyelocytes with CD 13,33 t(15;17)(q22;q12) More favorable if
azurophilic granules, CD2, +CD117 PML/RARa Variants responsive to ATRA
Auer rods all involve 17q12
AML with (t9;11) Monoblasts and CD 33,65,4, HLA-DR t(9;11)(p22;q23), Intermediate
promonocytes MLLT3-MLL
predominate
AML, therapy Multilineage dysplasia, CD 13, 33, 34, +CD 11q23 Less favorable;
related RS, increased basophils 56, 57 abnormality Median survival < 3
seen with years
topoisomerase
II
inhibitor-associated
AML
AML with t(6;9) Any morphology may be CD 13, 33, 38, DEK-NUP214; t(6;9) Less favorable
seen but HLADR, CD117 (p23;q35)
AML with inv(3) or myelomonocytic is most CD13, CD33, CD38, Inv(3)(121; q26.2) or Less favorable
t(3;3) common Any HLA-DR, CD117 t(3;3) (q21;q26.2)
morphology may be RPN1-EV11
seen except APL
AML with t(1;22) Megakaryoblastic CD41, CD61, square} t(1;22)(p13;q13) Less favorable
morphology CD13, CD33 RBM15-MKL1
with small and large
megakaryocytes
AML with FLT3 Any Any t(6;9)(p23;q34), Less favorable
mutation/duplication t(15;17)(q22;q12) or
normal
AML with NPM1 Myelomonocytic and CD13,CD33, CD34 is NPM1 mutation, More favorable in
mutation monocytic features negative cytoplasmic the absence of FLT3
expression
AML with CEBPA Variable, generally CD13, CD33, CD65, CEBPA mutation More favorable
mutation similar to less CD11b, CD15
differentiated AMLs in
FAB scheme (M1,2)
HEMATOLOGY 2

CYTOCHEMICAL STAINS: Cytochemical Reactions in Acute Leukemia

CYTOCHEMICAL CELLULAR ELEMENT BLASTS IDENTIFIED NOTES


REACTIONS STAINED
ENZYMATIC STAINS
 Enzymatic staining: the stain is not the enzyme. (e.g. CAE stain  enzyme is what we want to detect which is
inside in the cell) Purpose/Composition of stain: serve as a substrate to cause a reaction
Myeloperoxidase (MPO) Neutrophil primary Myeloblasts strong positive Differentiates ALL from
granules and auer rods Monoblast faint positive ANLL
For AML identification
(+) Activity: reddish brown
deposits (in cytoplasm of
granulocytes and
monocytes)

MPO enzyme deteriorates;


stain should be done only
on fresh specimens
Luekocyte Alkaline Present in tertiary granules Identify CML (leukemia; uncontrolled proliferation) from
Phosphatase of neutrophils leukemoid reaction (LR): transient
Reference Value: 30-185 LAP score

CML: WBC: >50,000/uL  LAP score: <0


LR: WBC: >50,000/uL  LAP score: Normal to High

Smear with LAP stain  observe microscopically  count


100 neutrophils  Grade

Kaplow’s LAP SCORE:


0 = no staining
+ = faint and diffuse staining
++ = pale and moderate amount of blue staining
+++ = strong blue ppt
++++ = deep blue or brilliant

Computation: Multiply the number of cell


by their grade e.g.
0 = 30  0
+ = 50  50
++ = 20  40
= 90 LAP score

Increased LAP Score:


(Controls: During the last trimester of pregnancy),
Polycythemia vera, Aplastic anemia, Hodgkin disease,
multiple myeloma, obstructive jaundice
Specific esterase Cellular enzyme Myeloblasts strong positive For AML identification
HEMATOLOGY 2

alpha-Naphthyl Marker for mature and


chloroacetate esterase immature neutrophil and
mast cells
(+) Activity: bright red
granules in cytoplasm
This enzyme is stable and
may last for months
Nonspecific esterase Cellular enzyme Monoblast strong positive For M4 and M5
alpha-naphthyl Acetate Marker for cells of
Esterase and alpha- monocytic origin
Naphthyl Butyrate Other cells: (+)
Esterase Megakaryocytes,
Platelets, Histiocytes,
Plasmacytes, some T-
lymphocytes
(+) Activity: red-brown/
dark red
Terminal deoxynucleotidyl Intranuclear enzyme Lymphoblast positive Present in the nucleus of
transferase (TdT) immature lymphocytes
primarily lymphoblast
Stain for ALL
Important in identifying
blastic transformation of
CML
e.g. CML  TdT  ALL CML
 MPO  AML
Tartaric Acid Resistant Acid For hairy cell leukemia(HCL) Other tartrate resistant Activity is indicated by
Phosphatase (TRAP) cells: sezary cells, purple to dark-red granules
histiocytes, T cell of acute in cytoplasm
lymphocytic leukemia
ACP: 5 isoenzymes
1-4: inhibited by tartaric
acid  NEGATIVE
5: resistant to tartaric acid;
inside lymphocytes of HCL
 positive to TRAP
Cyanide- Resistant For identification of Eosinophils (+) Activity: brown
Peroxidase Eosinophilic components
Acid Phosphatase Present in all hematopoietic Activity is indicated by
cells and found in purple to red granules
lysosomes Unstable and cannot be
stored
NON-ENZYMATIC STAINS
Periodic acid-Schiff (PAS) Glycogen Variable, coarse, block-like It stains glycogen,
positivity often seen in mucoprotein,
lymphoblast and glycoproteins, and high
pronormoblasts, molecular weight
myeloblasts usually carbohydrates by
negative although faint combining with the
HEMATOLOGY 2

diffuse reaction may aldehyde portion of the


occasionally be seen glycogen.

Positive in erythroblasts in
Di Guglielmo disease/
Erythroleukemia

(-) Activity: bright


fuschia pink (pattern
of staining varies with
each cell type)

Principle: positive in all


blood cells except normal
erythroblast but the
abnormal erythroblast in
M6 is positive in PAS.
For AML- M6 Identification

Gives a faint reaction with


lymphocytes in ALL;
Differentiates
ALL-L1 from ALL-L2 of ALL
 Both positive
L1: chunky or block-like
positivity of granules
Sudan Black B (SBB) Lipids and phospholipids Myeloblasts strong positive Differentiates ALL from
Monoblast faint positive ANLL
For AML identification
Gives the same
information as peroxidase
in the interpretation (+)
Result: dark purple-black
granules
Can be done on stored
specimens More sensitive
than chloroacetate in the
demonstration of mature
and immature neutrophils
and mast cells
Perl’s Prussian Blue Stains siderotic granules Use to detect hemosiderin
( for diagnosis of in urine
sideroblastic anemia)
Toluidine Blue O Binds with For recognition of basophils A metachromatic stain:
mucopolysaccharides in and mast cells gives different colors
blood cells (+) Result: Reddish Violet
HEMATOLOGY 2

ACUTE LYMPHOCYTIC LEUKEMIA o Secondary lymphoid organs: spleen and


 All is characterized by an uncontrolled growth of lymph nodes  needs antigenic
abnormal lymphoid cells. It is primarily a disease stimulation.
of childhood (young children) and adolescence, o Primary lymphoid organs: bone marrow
accounting 15% of childhood cancers and up to and thymus.
75% of childhood leukemia.  Common causes of death are infection (1st) and
 Peak: 2-5 years of age bleeding (2nd).
 Clinical Manifestations: The same with other  In children, the common causes of infections are
acute leukemias but onset (s/s) is more sudden. S. aureus, P. aeruginosa, C. albicans, H. influenza,
o In general, leukemia is characterized by and P. mirabilis  parasites are not that
anemia (decreased RBC), recurrent pathogenic but causes severe outcome
infection and bleeding disorders especially in immunocompromised individuals.
 Immunocompromised patients:
2 MAJOR CLASSIFICATIONS OF LYMPHOCYTES:
o Humoral Immunity: B cell ALL: signs
include lymphadenopathy. 1. B cells: humoral immunity  produces
Splenomegaly and hepatomegaly. antibodies
Eventual infiltration of malignant cells 2. T cells: cell-mediated immunity
into the meninges (lymphoblasts appear
NOTE:
in the CSF), testes, or ovaries.
Unlike neutrophils, it involves on defense but the
o Cell-mediated Immunity: T cell ALL:
activity is PHAGOCYTOSIS.
there may be a large mass in the
TOTAL NORMAL WBC COUNT: 4,500 -11,000/ 4.5-
mediastinum (near ribs) leading to 11.0x109/L Peripheral blood lymphoblast counts
compromised/abnormality of regional greater than 20 to 30 x 109/L
anatomic structures. More common in
teenage males. Laboratory Findings:
 Prognosis: depends on age at the time of  WBC count vary from low (<5.0 x 109/uL) to
diagnosis, lymphoblast load/tumor burden, high (>100 X 109/L)  poor prognosis; most
immunophenotype, and genetic abnormalities. are lymphocyte.
o The younger (common) the age the  Most abundant WBC: Neutrophil (60-80%); 
better prognosis. In adults  worse lymphocyte (20-40%);  monocyte 
prognosis eosinophils  basophils
o Immunophenotype: type of ALL: (L1, L2,  In ALL, predominant cell is immature
L3). lymphoblasts.
 BM is hypercellular/hyperplastic (red marrow;
 L3: most severe and have worst
has more hematopoietic cells uncontrolled
prognosis.
production of lymphocytes) and heavily
 L1: better prognosis among all
infiltrated with lymphoid cells.
the types. NOTE:
o T-ALL: bad prognosis ALL- most are lymphoblasts indicated by immature
o Worse outcome if the count of amount of basophilic cytoplasm
lymphoblast is too high (greater than 20 AML – most are myeloblasts
to 30x109/L)  poor prognosis: Hard to differentiate: Immature cells: larger in size,
hepatosplenomegaly, and large nucleus with basophilic cytoplasm  importance
lymphadenopathy all are associated of cytochemical staining and CD markers
with worse outcome (uncommon in differentiation.
AML)
HEMATOLOGY 2

mature
cells are
seen in this
type; negative
in CD34
T-ALL CD2, CD3, CD4, CD5, Rare and more
CD7, CD8, TdT common on
teenage
males.

WHO: Immunophenotyping

- Regardless of the classification: from CFU-L 


lymphoid antigens: CD34, TdT (nucleus of
lymphoblasts).
Further classification of B-ALL: Based on recurrent genetic
ALL IMMUNOPHENOTYP NOTES abnormalities: 7 types T-ALL not further classified
SUBTYPE E
Early CD34, CD19,
5% (less NOTE:
(pro/pre- cytoplasmic CD22,
common in CALLA: monoclonal COMMON ACUTE
pre) B-ALL TdT LYMPHOBLASTIC LEUKEMIA ANTIGEN: present on
children);
the leukemia cells of 70% of all patients; not present
11% (most
on normal peripheral lymphocytes but not specific
common in
for leukemia because it is also present on normal BM
adults)
cells that are positive for TdT and HLA-DR antigen;
Intermediat CD34, CD19, CD10, Common B-
only aids in identification of type II: intermediate B-
e (common) cytoplasmic CD22, ALL;
ALL
B-ALL TdT Common ALL FAB Classification of ALL: based on difference in
antigen morphology and cytochemical staining
(CALLA) are
Bases of FAB: Occurrence of individual cytologic
found  CD10
features (cell size; chromatin; nuclear shape, nucleoli,
has
degree of basophilia in the cytoplasm & presence of
relationship
cytoplasmic vacuolation); Degree of heterogeneity in
with CALLA
distribution among the leukemic cell population of
Pre B-ALL -CD34, CD19, 15%
some or all of those cytologic features
cytoplasmic CD22, (child);
Hand-mirror cells: lymphoblast that has
cytoplasmic m, 10%
(adults) contracted cytoplasm
TdT (variable)
 OIF= RBC  Neutrophils  Lymphocytes 
commo Monocytes  Eosinophils  Basophils
n in
children
less in
adults;
most
HEMATOLOGY 2

Criteria L1 L2 L3
(Burkitt type)
Cell size Small, homogenous Large, heterogeneous (large Large; homogenous
and small)
Nucleus Regular with occasional Irregular; clefting/indentation Regular, oval to round
clefting; Rare with occasional is common
clefting
Nucleoli Rare/small, inconspicuous Present (often large) 1-3 (vesicular)
Chromatin 1-3 (vesicular) Variable Finely stippled
Cytoplasm Scanty Moderate Moderately; vacuolation of the
cytoplasm
Basophilia Moderate Variable Intense
Incidence 15 y/o and below Older than 15 y/o Rarest to occur; Burkitt lymphoma
(ALL in t(8;14)
general is
more
common in
young
children (2-
5 y/o)
Most common and has the best Rarest and has the worst or
prognosis poorest prognosis;

NOTE: ALL resulting to gene fusion (BCR-ABL gene). Ph1 is


First stage of life: Infants and newborns: AML present in around 90% of CML adults, while
Young Children: ALL infants and children <2 years of age are often
Adults: CML Ph1 (-).
Older adults: CLL  Also known as Chronic Granulocytic Leukemia
Late/Last stage of life: Elderly: AML (CGL): most abundant cells is granulocytes
(neutrophils)  Disease of adults
 Philadelphia chromosome: Ph1 or Ph’  BCR-
CHRONIC MYELOPROLIFERATIVE DISORDERS ABL gene  CML; truncated Chromosome 22.
(CMPD) o Chromosome 9 is longer chromosome;
- CMPDs are a group of acquired, malignant chromosome 22 is shorter  both have
disorders that develop from the proliferation of p arm (short) and q arm (long)
an abnormal pluripotential stem cell. o Chromosome 9 (ABL gene is detached;
became longer) reciprocal translocation
1. CHRONIC MYELOGENOUS LEUKEMIA  Chromosome 22 (part is truncated in
(CML/CGL) BCR; but not detached; became shorter
 CML is a stem cell disorder commonly affecting due to truncation)  truncated
adult. Some patients show a chromosomal Chromosome 22 (Ph’): where new gene
abnormality called the Ph1 chromosome is formed: BCR-ABL gene  cause
(formed by the translocation of long arm of chronic leukemia  more oncogenic
Chromosome 22 to long arm of Chromosome 9 effect.
HEMATOLOGY 2

o Truncated chromosome 22: Ph1 has o CML: bone marrow defect; cells with
BCR-ABL gene expressed as mRNA (has abnormality  0 to low LAP score
genetic code); template for CHON o Neutrophils (Tertiary granules): with highest
synthesis  DNA transcription  and only cell with LAP activity  LAP is also
production of p210 protein: influence called Neutrophil Alkaline Phosphatase (NAP)
 chronic phase of leukemia  • Increased granulocytes: Basophilia; Eosinophilia;
and Monocytosis
excessive production of mature cells
o PPSC  CFU-GEMM  CFU GM  CFU-G
(granulocytic cells)
(granulocyte) and CFU-M (monocyte)
NOTE: • RBC and Plt. count decreased: Anemia and bleeding
disorder
• M:E = 10:1 or higher
o M:E– relationship between nucleated
granulocytic cells and precursor versus
precursors of erythrocytes
o In bone marrow, granulocytic cells is higher in
number than erythrocytes
o Normal ratio: 2-4:1
 Every 1 RBC there is 2-4 granulocytic
precursors
o In ALL, hypercullar BM but M:E ratio is not
affected or increased because what increases
are lymphoid cells
PROGNOSIS: Not all CML patients have Ph’

• Ph1 (+) patients have better prognosis than Ph1 (-)


Truncated chromosome 22: Ph1 has BCR-ABL gene  • Ph1 (+) : mostly Adults  CML; prognosis is better.
expressed as mRNA (has genetic code); template for • Ph1 (-) : Infants and <2 y/o  JMML or Monosomy 7
CHON synthesis  DNA transcription  production of under MSD/MPD
p210 protein: influence  chronic phase of leukemia o If you have abnormal chromosome the better
 excessive production of mature cells (granulocytic is your prognosis.
cells) o Effect of presence of Ph1 in CML  better
Imatinib mesylate: drug for CML which targets p210 prognosis
protein  produced as an effect of newly produced o If you have abnormal chromosome the better
BCR-ABL gene  Dominated by granulocytes  is your prognosis
neutrophils • High risk of transforming from chronic to blast 
Clinical Features: Splenomegaly; Hepatomegaly; Bone “blast transformation” or “acute exacerbation blast
pain crisis” is indicated by an increase of >20% blasts in the
PB or BM.
LAB FINDINGS: o If myeloblast fails to mature  acute
•WBC count: 50-600 X 109/L; ↓/0 LAP score leukemia; >20% blasts  can change in cell
•Basophilia; Eosinophilia; Monocytosis line due to release of abnormal chemicals
•Bone marrow: hypercellular with predominant and substances like cytokines that will
granulocytic cells (different stages of maturation); influence the differentiation of cell.
more red marrow  more hematopoietic cells) CML/CGL  ALL: TdT (+) AML: MPO (+)
• ↓/0 LAP score:
o Differentiated with leukemoid reaction (LR):
transient; cells with normal function 
normal to high LAP score.
HEMATOLOGY 2

Acute Leukemia Chronic Leukemia NOTE:


>20% are blasts/ <20% blast; more mature  Later stage  BM infiltrated with fibrous
immature cells  cells tissues  resembles and become
abnormal indistinguishable with BM myelofibrosis.
Fail to mature  More mature cells o Hallmark mark: teardrop cells in
prone to destruction myelofibrosis and later stage of
of blasts cell primary polycythemia vera.
Unlikely to become If blasts are destroyed  Hyperviscosity syndrome  increased cells
chronic  Can’t revert before maturing  can but few plasma volume  flow slower than
back to chronic progress to Acute Leukemia usual  increased pressure (to compensate)
leukemia o The slower flow  thrombotic
Poor prognosis Better prognosis condition.
NOTE:
Management: Repeated phlebotomy
2. POLYCYTHEMIA VERA  Prevent hyperviscosity of blood
 This is characterized by pancytosis/panmyelosis  Removes blood: Blood removed can’t be
due to clonal proliferation with an increase in all transfused  abnormal blood
cellular BM elements, specifically the red cell  Every blood volume removed; iron (60% in hb)
mass (RCM). is loss  possible develop Iron Deficiency
 Laboratory: Anemia (IDA).
o Generally lower EPO level
o High RBC count (>10M)
NORMAL POLYCYTHEMIA VERA
▪ Normal RBC count: 3.8-5.2
HCT = PCV x 100 2: Relative polycythemia
Millions TV vera
▪ Increased in all RBC 1: 50/100 x 100= 50% Decreased in plasma
measurement volume  changing the
o High Hct (>55% in males; >47% in total volume
females) 50/75 x100 = 66.67% 
o High Hb: >18.5 g/dL (M); >16.5 g/dL (F) increased hct
o High LAP, WBC count: 40-50,000/uL In here, bone marrow is
o High Plt count (>1000 x 109/L); more not defective but the hct
than 1 million is increased due to
reaction with underlying
▪ Normal plt. count: 150-
condition
450,000/uL
o High Basophils

WHO CRITERIA FOR THE DIAGNOSIS OF PV 3: True or Primary


polycythemia vera
 Elevated hb> 18.5 (M), 16.5 (F) g/dL, or other Plasma volume is the
evidence of increased Red cell volume. same however the PCV is
 Presence of JAK2 V617F or similar mutation such increased.
as AK2 exon 12 mutation. 75/100 x 100 = 75% 
increased hct
o Janus kinase gene  mutated gene 
dictates abnormal
Increased PCV  due to
production/proliferation and mutation increased production in
of cells. the bone marrow
(defective BM)
HEMATOLOGY 2

presence of
abnormal
hemoglobin

Inappropriate
response
Associated with some
kidney diseases

3. ANGIOGENIC MYELOID METAPLASIA/


IDIOPATHIC MYELOFIBROSIS/ MYELOFIBROSIS
WITH MYELOID METAPLASIA/ PRIMARY
MYELOFIBROSIS
- AMM is characterized by fibrosis,
megakaryocytic and granulocytic hyperplasia in
the bone marrow (marrow fibroblast).
- This is caused by an increase in defective
platelets as a result of dysplastic
megakaryocytopoiesis. These dysplastic cells
Polycythemia/Erythrocytosis: Characterized by an are prematurely destroyed resulting in the
INCREASED in HEMATOCRIT which may be due to: release of their a-granules that contain platelet-
1. A true increase in Red Cell Mass: PV/Primary derived growth factor (PDGF) which stimulates
polycythemia and secondary polycythemia fibrosis.
2. Just a decreased in plasma volume (relative o Fibrosis (marrow fibroblast)  caused
polycythemia) by dysplastic megakaryopoiesis 
DIFFERENTIATION abnormal megakaryocytes and platelets
Primary Secondary Relative  easily destroyed and lysed 
Polycythemia Polycythemia Polycythe released the contents of their granules
With bone No bone marrow mia
(alpha  PDGF (normally, promote
marrow defect defect No bone
vessel repair; abnormally, cause fibrous
marrow
defect tissue proliferation  scar formation
This is the Appropriate/compens Associated and dense)
myeloprolifera atory with a - Blood film shows predominance of teardrop
tive disease  Characterized decrease cells. Marrow fibrosis often result to “dry tap”
Absolute type by an increase in the during bone marrow aspiration.
Belongs to in the RCM plasma - Bone marrow failure to produce blood 
CMPD &Hct due to volume increased in fibrous tissue and depleted blood
some with no cells; what will produce are liver and spleen that
identifiable increase in has residual function for hematopoiesis
cause RCM & however there would also be fibrosis in these
 High in RCM is EPO organs especially in the spleen.
secondary to
- With increased blood cells and fibrous tissues
high in EPO
- EXTRAMEDULLARY FIBROSIS: fibrosis in the
 Hypoxia: can
be seen in liver and spleen
high altitude, - Middle aged to older people; rare to occur in
high O2 children
affinity and - Bone marrow: 2 normal tissues
HEMATOLOGY 2

o Yellow: Marrow fats - Spontaneous aggregation even in the absence


o Red: Hematopoietic marrow of vessel injury  Megakaryocyte proliferation
- Blood film shows predominance of teardrop with large and mature morphology; no or little
cells. granulocyte or erythroid proliferation.
o PBS: teardrop cells  hallmark finding; - Platelet aggregation studies show platelets have
indicates the disease decreased/abnormal aggregation with
o Formation of teardrop cell can be epinephrine.
medullary (BM) and extramedullary - In vivo aggregation  platelets tend to obstruct
(SPLEEN) circulation
o Due to narrow circulation in the fibrotic o Two types of thrombosis:
BM and spleen  RBCs squeeze ▪ Arterial: involves platelets 
themselves  deform in order to pass arterial vasoocclusion by
through or enter the limited platelets aggregates 
circulation severe damage in cell decreased circulation and
membrane  retained the shape  oxygenation  called ischemia
TEARDROP CELL ▪ Venous: involves fibrin
- During bone marrow aspiration/core Biopsy 
University of Illinois needle targeting marrow o Arterial or venous thrombosis: 
sinusoids where bone marrow sample is Transient Ischemic Attacks (e.g.
aspirated. priapism (penis); digital ischemia
o Marrow fibrosis often result to “dry (peripheral tissues (digits: toes and
tap” during bone marrow aspiration. finger))
o DRY TAP: difficulty in aspirating that is ▪ More on arterial  decreased
very painful to the patient oxygenation
o Bone marrow should be spongy but due
▪ Toe ischemia: bluish coloration
to fibrous tissues infiltrated it becomes
on the toes
scarred/hard  hard aspiration.
▪ Priapism is a prolonged
erection of the penis. The
4. ESSENTIAL/PRIMARY THROMBOCYTOSIS
persistent erection continues
- ET characterized by thrombocytosis with
hours beyond or isn't caused by
spontaneous aggregation of abnormal platelets.
sexual stimulation. Priapism is
Platelet aggregation studies show platelets have
usually painful. Although
decreased aggregation with epinephrine.
priapism is an uncommon
- 2008 WHO criteria: platelet count of
condition overall, it occurs
>450x109/L. Megakaryocyte proliferation with
commonly in certain groups,
large and mature morphology; no or little
such as people who have sickle
granulocyte or erythroid proliferation; does not
cell anemia.
meet WHO criteria for CML, PV, IMF, MDS, or
▪ Priapus: minor fertility god in
other myeloid neoplasm; demonstration of
Greek mythology, who was also
JAK2 (V817F) mutation or other clonal marker,
the protector of livestock, fruit
No evidence of reactive thrombocytosis.
plants, and male genitals. He
- PRIMARY: main defect is production  bone
was depicted as having an
marrow defect  cancer of BM  excessively
oversized and permanent
produce thrombocytes
erection.
- SECONDARY: in reaction, also known as
reactive thrombocytosis
HEMATOLOGY 2

NORMAL SMEAR ABNORMAL SMEAR (Essential thrombocytosis)

Platelet count: 8-20/OIF that


contains 200 RBCs.

ET/ PRIMARY THROMBOCYTOSIS POLYCYTHEMIA VERA


Increased platelets (ONLY) with NO increased in RCM Increased in all blood cells with prominent increased RCM
and other blood cells
JK mutation gene JK mutation gene
PRIMARY THROMBOCYTOSIS SECONDARY/REACTIVE THROMBOCYTOSIS
Platelet count often exceeds 1M/uL or >1000 x109/L Platelet count is high more than 450,000/uL but rarely
exceeds 1M/uL or >1000 x 109/L
Not reactive: Increased in the production by defective Platelet pool: 1/3 spleen and 2/3 circulation
bone marrow  JAK2(V617F) mutation Reactive: When 1/3 platelet in the spleen is mobilized into
the circulation  reactive thrombocytosis (transient)
With permanent damage, unless treated with bone
marrow transplant

Abnormally produced due to defective marrow  Normally produced (from spleen; just mobilized) 
Abnormal platelet function normal platelet function
HEMATOLOGY 2

CHRONIC LYMPHOPROLIFERATIVE DISORDERS NOTE:


1. CHRONIC LYMPHOCYTIC LEUKEMIA (CLL) CLINICAL SIGNS AND SYMPTOMS INCLUDE:
 CLL is characterized by abnormal proliferation  Enlarged lymph nodes (lymphadenopathy),
of lymphocyte that are unresponsive to spleen (splenomegaly), and liver
antigenic stimuli. CLL is less likely to undergo (hepatomegaly)  CLL will more likely show
blastic transformation but CLL patients are these S/s over CML.
predisposed to develop autoimmune hemolytic  Pruritus due to recurrent skin infection (often
with Herpes zoster).
anemia (AIHA)
LAB FINDINGS:
o Less likely to undergo blastic
Lymphocyte count: 10-150 X 109/L  very high and
transformation
mature
▪ does not become ALL or AML  More on lymphocytes: thin rim cytoplasm;
o Dormant B-cells: unresponsive to Ag morphologically normal but is fragile 
stimulation and treatment during smearing  smudge cell  CLL
▪ B cells are stimulated by ▪ Differentiate smudge cells from
antigens  transform into basket cell  nuclear remnants of
plasma cells  produce granulocytes; seen in chronic
antibodies. granulocytic leukemia (CGL/CML)
▪ Decreased IgG production: IgG ▪ Smudge cells: formed IN-VITRO 
(2nd; produce at chronic phase, caused by manner of smearing; does
but once produce it continues not seen in wet preparation.
 Lymphoblasts: larger and basophilic
 confers long term or lifetime
cytoplasm
protection but in here, it is
Many smudge cells on the blood smear
decreased thus patients prone Hypogammaglobulinemia.
in reinfection). TREATMENT/MANAGEMENT:
▪ Affected B-cells does not 1. Leukapheresis
produce normal IgG but  Apheresis: separation technique for
produces abnormal ones that is blood components  Whole blood:
directed against the patient’s PRBC, Plasma (PPP and Fresh plasma,
RBC leading to AIHA. cryoprecipitate and cryosupernate),
▪ Warm Autoimmune Hemolytic Platelets (plateletpheresis), and WBC
Anemia (WAIHA)  IgG – through leukapheresis  separates
into Granulocytes and/or
related: warm type
lymphocytes
▪ Recurrent skin infection: 2. Regular injection with gamma-globulins (IgG) 
Herpes zoster or Shingles  manage infection.
pruritus
▪ Chickenpox: initially appears as
papules on the skin and when 2. HAIRY CELL LEUKEMIA/ LEUKEMIC
reoccurs it is now called herpes RETICULOENDOTHELIOSIS
zoster  HCL shoes presence of small lymphocytes with
▪ IgM (1st; peaks immediately many fine cytoplasmic extensions.
but declines rapidly  does not o It affects B-cells with presence of ACP 5
confers lifetime protection; cold o Microscopically, cells have cytoplasmic
type AIHA). hair-like protrusions
 PROGNOSIS: 3-4 years  Prognosis is quite good (benign), and can be
longer than 10 years.
o Benign and has a good prognosis
HEMATOLOGY 2

 Both CLL and HCL  more common in older o Histological finding: accumulation of
adults (>50 y/o) and more on males lymphocytes called  Pautrier’s
 Pancytopenia  unique finding and important microabscess in epidermal biopsy
feature of HCL.  SEZARY SYNDROME: Flow through lymphatic
circulation and reach lymph
NOTE: o Blood smear: Sezary cells  lymphoid
 Clinical sign and symptoms: splenomegaly is cells with convoluted or irregular
most consistent (splenectomy is often
nucleus
considered).
 Laboratory diagnosis: Cytochemical stain  NOTE:
TRAP (+) MYCOSIS FUNGOIDES
o Isoenzyme 5 of ACP  unique  This affects the mature T cells. This is an
because it is resistant and not early stage of Sezary syndrome and is
inhibited by Tartaric acid  when characterized by pruritus, eczematoidal
employed with ACP stain  ACP 1-4 psoriasis form non-specific exfoliative
(negative); ACP 5 (positive) dermatitis. Diagnostic evaluation of the skin
reveals Pautrier’smicroabscesses (clusters of
lymphocytes forming on the skin)
accompanied by parakeratosis.
3. PRO-LYMPHOCYTIC LEUKEMIA
 EARLIER STAGE
 CLL is characterized by an increase in
prolymphocytes, with almost total replacement SEZARY SYNDROME
of the bone marrow. This has a poorer  This stage manifests infiltration of the lymph
prognosis than HCL and CLL. nodes and viscera, characterized by band-like
 RARE BUT MOST SEVERE infiltrates of lymphocytes with cerebriform
 PROGNOSIS: poor (<1 year)  life expectancy nuclei called Sezary cells.
if left untreated.  ADVANCE STAGE

NOTE: PLASMA CELL DYSCRASIAS


LAB FINDINGS: Paraproteinemia: MM and WM  malignancies
 HIGH LYMPHOCYTES WITH PREDOMINANCE involving plasma cells.
OF PROLYMPHOCYTES  less mature  Excessive proliferation of plasma cells even in
o STAGES of lymphocyte maturation: the absence of antigenic stimulation 
LYMPHOBLAST  PROLYMPHOCYTE excessive production of abnormal
 LYMPHOCYTE immunoglobulins.

1. MULTIPLE MYELOMA
OTHER LYMPHOCYTE and PLASMA CELL NEOPLASMS  This is the most common clonal proliferative
DIFFERENT STAGES OF A SINGLE NEOPLASTIC malignancy of plasma cells.
DISORDER  Monoclonal gammopathies  abnormal cells
produce a single type of immunoglobulins.
 Both affects T-cells
 Of the cases, approximately 50% produce only
o T-helper cell (lymphatic tissue)  skin:
IgG; 20% only IgA; and 15% only the L-chain
MYCOSIS FUNGOIDES
o Plasma cells: 50% - only IgG; or 20% -
o SKIN: affected T-cells will cause damage
only IgA and/ or 15% - only L-chain 
in the epidermal layer  parakeratosis
interfere in platelet function
and pautrier’s microabscess. Skin
become scale that causes eczematoidal  Marrow infiltration often leads to bone pain.
or fungal lesions. o Myeloma cells: cancerous and leukemic
plasma cells that cause increased
HEMATOLOGY 2

production of antibodies  tumor has sialic acid in their


formation in bone marrow  lytic bone phospholipid layer that
lesions  bone pain imparts a negative charge)
NOTE: ESR: 3 stages
 Initial Rouleaux: first 10 mins
Clinical sign and symptoms:
 Lytic bone lesions  rouleaux  becomes
 Hyperviscosity: due to abnormal proteins in heavier
the plasma  Rapid Settling: succeeding 40
o Causes hypertension and mins  larger sediment will
Cardiovascular Disease (CVD) settle faster
 Bleeding disorders
 Final Settling: last 10 mins
o Platelets and endothelial cells are
coated with the abnormal proteins o Hypercalcemia: due to abnormal
 preventing the expression of metabolism of calcium  bone
glycoproteins and other receptors  lesions and pain
abnormal adhesion and aggregation BLOOD FILM FINDINGS
Laboratory findings reveal:  Flame cells: plasma cell with red to pink
o Chemistry: Bence Jones Protein (BJP) cytoplasm associated with increase in IgA
o Myeloma cells may produce BJP  o Plasma cells with cytoplasm filled
part of immunoglobulin but with IgA
represent only the L-chain  Low o Appear like a flickering candle
MW  easily passes through
glomerulus  seen in urine 
urinary screening
o Urine screening: unique solubility
characteristic  when heated at  Dutcher bodies: intranuclear crystalline
certain temperature it can structures of abnormal IgG.
precipitate and be dissolved o Contains precipitated IgG in the
 Not heated or <40°C = clear/ nucleus of the plasma cells
soluble
 40-60°C = urine becomes
turbid due to precipitation
of protein; BJP  insoluble
 100°C = clear; BJP  soluble  Grape cell/Mott cell, Morula cell: plasma cell
o Urinalysis: many hyaline casts and other that contains small colorless or blue/pink
casts and positive BJP protein vacuoles that appear like grapes
o Hematology: o Plasma cells whose cytoplasm are
o Elevated ESR: Rouleaux formation filled with several protein globules 
 Because of the abnormal russel bodies: are globules seen in
proteins produced, zeta the cytoplasm containing proteins
potential of the RBC is and IgG  made the cell look like a
altered  rouleaux bundle of grapes
formation (normally in vivo o Russel bodies: accumulations of IgG
RBC does not form into
rouleaux  repel each other
through zeta potential; RBC
HEMATOLOGY 2

LYMPHOMAS
- Lymphomas are cancers of the lymph nodes
characterized by uninhibited growth of cellular
 Rouleaux formation elements normally found in lymphatic tissues
resulting to lymph node enlargement.
o Lymph nodes are well-organized
because they are involved in the
immunity.
- Proliferative disease affecting some of the blood
forming cells and tissues outside the bone
2. WALDENSTROM MACROGLOBULINEMIA marrow  lymph tissues: lymph nodes and
 This is the second most common type of plasma vessels
cell dyscrasia. - May affect either B-cells or T –cells
 Almost similar with MM
 Myeloma plasma cells are more involve in the 1. NON-HODGKIN LYMPHOMA (NHL)
production of IgM.  A more common type than Hodgkin lymphoma.
o Malignant plasma cells show increase It is a B-cell lymphoma characterized by painless
production of IgM (>3 g/dL) lymph node enlargement (cervical nodes are
o IgM: pentamer; big  most often involved)
macroglobulinemia  Cervical nodes  GIT lymph nodes 
 Also called Lymphoplasmacytic lymphoma  enlargement.
can affect lymphocytes that are not transformed  Represent 60% of lymphoma cases
into plasma cells.  CAUSES: congenital immunodeficiency diseases
 Hyperviscosity: due to macroglobulins and viral agents
o Since it is a cancer, mechanism isn’t
NOTE: clearly defined.
LAB FINDINGS:  CLASSIFICATION:
o Increased IgM (>3 g/dL)
o Low grade: enlargement is slow; can
o Rouleaux formation  Increased ESR
take years for the enlargement of lymph
o BJP can be produced
nodes to be diagnosed as one
DIFFERENCE BETWEEN WM AND MM
1. WM: predominantly IgM o Intermediate grade: Rapid enlargement
MM: predominantly IgG/IgA/L-chain o High grade: More rapid enlargement

2. MM: affects B-cells and plasma cells precursors 2. BURKITT HODGKIN LYMPHOMA
WM: only plasma cells are affected  This is a B-cell neoplasm associated with EBV
and HIV infections. It is endemic among African
children (observed as jaw mass). Cytogenic
RARE CONDITIONS:
abnormality involves a translocation of C-myc
3. PLASMA CELL LEUKEMIA gene on chromosome regions (Ch8:14)
 >2x109/L plasma cell count  One type of non-hodgkin lymphoma which can
be B-type or T-type  can affects both B-cells
4. HEAVY CHAIN DISEASE (HCD) and T-cells
 This shows an abnormal synthesis of heavy  Lymphoma that when it infiltrates the bone
chains (Gamma HCD; Alpha HCD) marrow  it becomes leukemic  ALL-L3
 ALL-L3 and Burkitt  related to EBV and HIV
 Problem: translocation of a gene between 8 and
14 (more common; written as t (8;14)) or 8 and
HEMATOLOGY 2

2 or 8 and 22; the gene translocated is is C-myc  Hallmark finding: REED-STERNBERG (R-S) CELL
gene or LACUNAR HISTIOCYTES
o C-myc gene is a normal gene that both
regulates proliferation and apoptosis
but its translocation will cause it to be
abnormal thus it will become
overexpressed  abnormal function
 Prominent feature: rapid proliferation of
affected cell  rapid enlargement of lymph  Giant lymph cell that is usually 4-8x larger than
nodes lymphocytes.
 DIAGNOSIS: Lymph node biopsy: diffused  Giant multi/binucleated: more common and
lymphoid proliferation (many lymphoid cells; each half is identical to each other
monocyte and macrophage: clears cellular o Nuclei  shows distinct nucleolus with
debris)  STARRY SKY. very prominent nuclear halo and distinct
nuclear envelope  giving owl-eyes
image
 When cut in half; almost identical  MIRROR
IMAGE CELL.
 When seen is pathognomonic of the disease
 CBC is not the appropriate procedure because it
is not a cancer of the bone marrow thus the
disease have a normal CBC.
NOTE:  But in cases wherein the lymphoma becomes
WHO Classification: leukemic; CBC can be included.
Endemic: more common in African children   LEUKEMIC PHASE OF LYMPHOMA: CBC 
enlarged jaw or with jaw mass or upper lymph nodes Leukocytosis and lymphocytopenia are observed
Sporadic: more common in Unites States 
in advance cases  poor prognosis.
American  begins at GIT  enlargement of GIT
lymph nodes TWO CRITERIA:
Immunodeficiency: EBV and HIV
A. ANN HARBOR CLASSIFICATION OF HIGKIN
LYMPHOMA
3. HODGKIN LYMPHOMA  Region of RN (I-IV):
 This is characterized by painless enlarged lymph o Suffix A: no s/s
nodes usually in the neck and/or in some, the o Suffix B: with s/s
cervical nodes that spreads in an orderly fashion. o Subscript E: indicates involvement of
o If enlargement is rapid (severe cases)  extra lymphatic tissue
usually is painful. o Subscript S: indicates involvement of
o Orderly fashion enlargement  it the spleen.
follows lymphatic circulation  Stages:
 More on B cell lymphoma o Stage I: only one lymph node region
 Signs and symptoms include: affected
o GENERAL: fever, night sweats, or 10% o Stage II: two or more lymph node
unexplained weight loss in 6 months, or region affected but all located in one
a combination of both. part of the diaphragm either upper or
 Diagnosis: lymph node biopsy  SPECIFIC lower.
IDENTIFICATION
HEMATOLOGY 2

o Stage III: two or more lymph node


region affected but are disseminated in
both parts of the diaphragm
o Stage IV: involvement of an extra
lymphatic tissue (spleen, liver or any
other organs)

B. RYE CLASSIFICATION (WHO)


 based on extent of lymphocyte infiltration and
the abundance of R-S cells
a. Lymphocyte Predominant: Abundant
Lymphocytes with normal and abnormal
forms; no fibrosis; with few R-SCs
b. Nodular Sclerosis: Moderate Lymphocytes
with nodulating collagen bands; R-SCs in
clear zones
c. Mixed Cellularity: Moderate lymphocytes in
diffuse pattern
d. Lymphocyte Depleted: few lymphocytes
with diffused fibrosis and many R-SCs

Stage Rye Histological Prognosis


Classification Features
I Lymphocyte Lymphocytes BEST
predominant and R-SCs
II Nodular Nodules of GOOD
sclerosis lymphoreticu
lar cells and
lacunar RSCs
III Mixed Mixture of FAIR
cellularity lymphocytes,
eosinophils,
plasma cells
and RSCs
IV Lymphocyte Lymphocytes POOR
depleted and RSCs
HEMATOLOGY 2

FINALS o Proper position of patient during


phlebotomy: in comfortable position,
whether upright/sitting or lying in bed.
ERYTHROCYTE DISORDERS
o Sudden change in position:
 ERYTHROCYTIC DISORDER: affects RBC
Hemodilution: upright to lying: shift of
functions  oxygen transport as conducted by
fluid from extravascular to
its hemoglobin content thus affecting the oxygen
intravascular/circulation  decreased
delivery function of RBCs called ANEMIA.
RCM.
 ANEMIAS: are group of clinical conditions o Hemoconcentration: lying to upright:
associated with either a decrease in the number shift of fluid from intravascular to
of total red blood cells, a decrease in the
extravascular increased RCM (PV) 
concentration of hemoglobin, and/or a decrease REMEDY: wait for at least 20 minutes to
in the hematocrit (in any or all of the RCM) in
elapse for the fluid to equilibrate
relation to the individual’s age, gender and o IV fluid: hemodilution
geographic location.
o Anemia conditions can be absolute or CLINICAL SIGNS:
relative.
General signs and symptoms due to lack of oxygenation
o Decrease in RCM:
in tissues
 Hemoglobin (single lab test
important in screening anemia)  Easy fatigability and dyspnea on exertion  lack
 RBC count of oxygenation
 Hematocrit  Vertigo and faintness  drop in circulation thus
 Decrease oxygen delivery in less circulation and oxygenation in the brain
tissue preventing normal o Nervousness/faint  REMEDY: patient
cellular respiration. must lay down improve circulation in
 POLYCYTHEMIA VERA: all findings are opposite the brain
of anemia  increase in all  increase in RCM  Palpitation, rapid bounding pulse, systolic
(absolute PV) or decrease murmurs and headache o Due to lack
plasma/hemoconcentration (relative PV)  oxygenation  systolic murmur  heart
hematocrit (single lab test important in compensates
screening PV) o Brain demands large amount of oxygen
therefore the heart compensates by
TYPES:
redistributing the circulation of blood
 ABSOLUTE: True decrease in RCM  Decreased through transporting more oxygen to
in production (bone marrow defect that does not the brain causing an increase in cardiac
release a normal RBCs); increased loss output.
(hemorrhagic); increased destruction o Compensation of heart: increase cardiac
(hemolytic) output  increase pressure
o Normal plasma volume but PRBCs or o Low Blood Pressure due to decrease
PCV is decreased blood output causing increase cardiac
 RELATIVE: related to volume of plasma: plasma output causing increase pressure
expansion  hemodilution: shift of fluid from leading to headache  Pallor (lacks
extravascular to intravascular; false hemoglobin  heme: gives the normal
interpretation: decreased in cell count, hb and red pigment of blood)
hct.  low BP (lack of blood)
o Specimen collection: Very important to  Slight fever (especially when coexistent with an
consider infection) and dependent edema
HEMATOLOGY 2

SPECIFIC CLINCAL SIGNS

JAUNDICE: yellow discoloration of the LEG ULCERS: open wounds most KOILONYCHIAS: spooning of nail bed
skin, eyes, nail bed due to commonly seen lower extremities  seen in Iron
accumulation of unconjugated because it is affected by thrombosis Deficiency Anemia
bilirubin (persist in circulation(yellow  in sickle cell anemia  rigid cells
plasma); water insoluble and bound  hyperviscosity of blood leading to IDA  hookworm infections
to albumin; not excreted in the urine) thrombosis Iron is very much needed by the body:
 specific sign seen in hemolytic hemoglobin and other heme
anemia In normal condition: RBCs are containing CHON (myoglobin and
discocyte, flexible and deformable  cytochrome) and tissues  heme
pass through small vessels. contains iron that give offs normal
In sickle cell anemia, rigid cells and color of blood and flesh.
not deformable therefore they
obstruct the circulation leading to
hypervicosity of blood  thrombosis
 vasoocclusion decrease in
oxygenation  delay wound healing

FRONTAL BOSSING: seen in NEUROLOGIC DEFICIT: seen in CYANOSIS: seen in


Thalassemias: Betathalassemia major Vitamin B12 deficiency  decrease methemoglobinemia  increase in
(more common) and DNA production leading to abnormal hemoglobin: hemoglobin
hemoglobinopathies megaloblastic anemia (affects blood Hemoglobin “M”s  methemoglobin:
cell maturation) and destruction of hb that is not protected from
Cause: Skull (flat bone) that is active in CNS tissues due to inefficient myelin oxidization  easily oxidized (ferrous
hematopoiesis. In B-thalassemia sheath  severe cases: mood swings to ferric form)  Hb M Boston, Hb M
major: no hemoglobin(A1) which is and change in behavior  Iwate, Hb M Hyde park, Hb M
the major hemoglobin in adults so the Megaloblastic madness Saskatoon, Hb M Milwaukee, Hb M
blood production tries to compensate Osaka and Hb M Fort Ripley.
 red marrow expansion inside the Vit. B12: important and serve as a Methemoglobin is normal in blood
flat bone producing more blood cells cofactor in many metabolic reaction: but in small amounts: >3.5% but if
 bone structure also expands  DNA synthesis and maintenance of increased it becomes abnormal
skeletal deformity myelin sheath of the brain and spinal because it cannot transport oxygen
B-THALASSEMIA: manifest mongoloid cord or CNS tissues especially in distal tissue like fingers.
features due to skeletal deformity
HEMATOLOGY 2

COMPENSATORY MECHANISMS
PHYSIOLOGIC RESPONSE HEMATOLOGIC RESPONSE

Immediately occur Take time to occur


. Shift to the right of the Oxygen Dissociation Curve Erythropoiesis: 3-5 days (from pronormoblast to
Y-axis = oxygen saturation; increase affinity of hb to O2 reticulocyte)
X-axis = partial pressure of oxygen; decrease affinity of  Normally: 3.5 days will be spent in BM and 1 day
hb to O2 in the circulation mature RBC
 Anemia: reticulocyte will be released in the
2.Increase 2,3 DPG shift to right in ODC and more circulation less than its normal (<3 days); if the
oxygen will be released towards the tissue number of days in the BM is shorten, the longer
TWO FORMS OF HEMOGLOBIN BASED ON the number of days the reticulocyte
OXYGENATION: will take in the circulation
1. Oxyhemoglobin – “R” relax form; No 2,3 DPG; Increase in erythropoietin (EPO)
fully oxygenated  increased affinity to  Increase erythroid precursors
oxygen; but once in tissue there will be  Accelerates rate of proliferation and maturation
decreased in the affinity 2,3 DPG inserts in the
 Accelerate release from the BM to the PB
hb
Increase shift cells/stress reticulocyte
2. Deoxyhemoglobin – “T” tense form; with 2,3
DPG that lowers the hb affinity to oxygen
release oxygen in the tissue
3.Selective redistribution of blood flow to areas of
highest oxygen demand like in the brain

4.Increased cardiac output

RETICULOCYTE SHIFT/ STRESS CELLS/ RETICS


 Immature cell  Prematurely released
 Normal count: Adults: 0.5-1.5% (average: 1%  More immature than reticulocyte
Retics)  Increased RNA
HEMATOLOGY 2

o Maintains 1% retics everyday o In anemia, prolonged in the circulation 


o In anemia, prolonged in the circulation increased retics count; and if they are shift
 increased retics count; and if they are (more immature more RNA)
shift (more immature more RNA)
Romanowky stain – polychromatic Supravital stain – bluish granules More granules  shift cells

MAJOR CAUSES OF ANEMIA o D. latum - tapeworm that lives in ileum;


1. GENETIC FACTORS interferes in the absorption of Vit. B12
- Abnormalities in the structure of some anemia leading to Vit. B12 deficiency
molecules present in the RBC. anemia or fish tapeworm anemia; often
a. Hemoglobin C, S: can’t function well in referred as bothricephalus pernicious
delivering oxygen leading to anemia. anemia
b. Cell membrane: abnormal spectrin and o Hookworms – Ancylostoma duodenale
ankyrin due to abnormal gene  RBC easily and Necator americanus: blood sucking
deformed  spherocytes/elliptocytes) nematodes that attaches in the mucosal
c. Enzymes: affects metabolism of RBC (like of the small intestine through their
G6PD deficiency  needed in hexose buccal cavity. As they move to one area
monophosphate shunt ) to another, the area of attachment will
be left behind as chronic/minute
2. ENVIRONMENT FACTORS bleeding area and mechanical sucking it
- Factors that interfere with the normal will cause blood loss (hb loss  iron
development and functioning of RBCs. loss especially chronic infection with
a. Lead exposure may lead to Sideroblastic heavy worm burden  IDA)
Anemia (SDA); lead interfere in heme o Malaria/Plasmodium spp. and Babesia
synthesis affecting the utilization of iron  – live inside the RBC  ruptures and
iron cannot be utilized therefore precipitate destroy RBCs hemolysis  hemolytic
as siderotic granules anemia
b. BENZENE – laboratory reagent that exposure
may lead to developing aplastic anemia; 4. IATROGENIC FACTORS
toxic and damages BM thus preventing - These are results of side-effects of legitimate
production of blood cells medical treatment/procedures that may
c. Failure to acquire nutrients, vitamins, interfere with the normal development and
minerals (e.g. from vegetables)  function of RBC
Nutritional Deficiency Anemia: Iron a. Prolonged use of antibiotics:
deficiency (IDA), Lacks dietary vegetable and Chlorampenicol – destroys BM leading to
meat (poultry product)  B12 and Folate development of aplastic anemia b. Surgery
Deficiency – hemorrhagic/blood loss anemia

3. PATHOLOGICAL FACTORS
- infectious organisms and parasites
a. PARASITES
HEMATOLOGY 2

LABORATORY DIAGNOSIS NOTE:


- To screen for the presence of anemia  Reference Value:
performs CBC  Normocytic: 80-100 fL
1. HEMOGLOBIN (ANEMIA) AND HEMATOCROT  Microcytic: <80 fL
(PCV)  Macrocytic: >100 fL
 Single test to screen anemia: Hemoglobin  if FORMULA:
low, proceed with stated test below. 𝑯𝒄𝒕 (𝑳/𝑳) × 𝟏𝟎𝟎 𝑯𝒄𝒕 (%) × 𝟏𝟎
𝒐𝒓
𝑹𝑩𝑪 (𝟏𝟎𝟏𝟐 /𝑳) 𝑹𝑩𝑪 (𝟏𝟎𝟏𝟐 /𝑳)
NOTE:
HEMOGLOBIN
 Reference Value:  MCHC: refers to average Hb concentration of
FORMULA: red cells in a given volume of blood
𝑨 𝒐𝒇 𝒕𝒆𝒔𝒕 (𝒖𝒏𝒌𝒏𝒐𝒘𝒏)
NOTE:
𝑨 𝒐𝒇 𝒔𝒕𝒂𝒏𝒅𝒂𝒓𝒅
Reference Value:
× 𝑺𝒕𝒂𝒏𝒅𝒂𝒓𝒅 𝒄𝒐𝒏𝒄. (𝟏𝟓𝒈𝒎/𝑳)
= 𝒈/𝑳 𝒐𝒓 𝒈/𝒅𝑳 (𝒘𝒊𝒕𝒉 𝒅𝒆𝒎𝒊𝒄𝒂𝒍)  Normochromic: 320-360 g/L or 32-36% or 32-
HEMATOCRIT 26 g/dL
 Reference Value:  Hypochromic: <32%
 Male: 140-175 g/L  Spherocytic  full of Hb: >36%
 Female: 123-153 g/L FORMULA:
𝑯𝒈𝑩 (𝒈/𝒅𝑳) 𝑯𝒈𝑩 (𝒈/𝒅𝑳) × 𝟏𝟎𝟎
 At birth: 150-200 g/L 𝒐𝒓
FORMULA 𝑯𝒄𝒕 (𝑳/𝑳) 𝑯𝒄𝒕 (%)
1. INDIRECT:
𝑯𝒄𝒕 = 𝑷𝑪𝑽 × 𝑹𝑩𝑪 𝑪𝒐𝒖𝒏𝒕
3. RETICULOCYTE COUNT; RETICULOCYTE
2. DIRECT
𝑯𝒕 𝒐𝒇 𝑷𝑪𝑽 × 𝟏𝟎𝟎 PRODUCTION INDEX (RPI) OR SHIFT
= 𝑽𝒐𝒍. 𝒐𝒇 %𝑯𝒄𝒕 CORRECTION
𝑯𝒕 𝒐𝒇 𝒘𝒉𝒐𝒍𝒆 𝒃𝒍𝒐𝒐𝒅 (𝑻𝑽)
𝒐𝒓  Compensatory mechanism of BM is to increase
𝑷𝑪𝑽 its reticulocyte production  increased
= 𝑳/𝑳 (𝒅𝒆𝒄𝒊𝒎𝒂𝒍)
𝑻𝑽 reticulocyte count.
 RPI: to measure whether the increase in
production is parallel or compensatory to the
2. BLOOD INDICES: MCV, MCHC, RDW
degree of anemia
 For morphological classification of anemia
o Index calculated to correct for the
 RDW: represents the ratio of the standard
presence of shift reticulocytes that
deviation to the MCV )Width of histogram)
otherwise may falsely elevate the visual
o Index of relative anisocytosis; may also
reticulocyte count
indicate poikilocytosis
o Increased in hemolytic and hemorrhagic
REFERENCE VALUE: anemia
 11.6- 14.6% o Reference value: 1 if hct is 0.45
 >14.6% indices variation in sizes
NOTE:
RETICULOCYTE COUNT:
 MCV: refers to the average volume of PCV of the  Reference value: 0.5 -1.5%  average: 1%
individual red cells in a given blood samples.
o It is used as an estimation of the average RETICULOCYTE PRODUCTION INDEX (RPI)
size of the RBC  Reference value: 1 if hct is 0.45
𝑪𝒐𝒓𝒓𝒆𝒄𝒕𝒆𝒅 𝒓𝒆𝒕𝒊𝒄𝒖𝒍𝒐𝒄𝒚𝒕𝒆 𝒄𝒐𝒖𝒏𝒕
o No dependable when RBC vary markedly  FORMULA:
𝑴𝒂𝒕𝒖𝒓𝒂𝒕𝒊𝒐𝒏 𝒕𝒊𝒎𝒆 (𝒅𝒂𝒚𝒔)
in size.
HEMATOLOGY 2

4. MYELOID: ERYTHORID RATIO B. ANEMIA ASSOCIATED WITH DEFECTIVE HGB


 Ratio between granulocytes and erythrocytes SYNTHESIS
precursor inside the BM  Defect in globin or heme synthesis
 Normal ratio: 3:1  1 RBC precursor: 3  Heme: IDA, SDA
Granulocytic precursors  Globin: thalassemia
 CGL/CML ratio: >10:1  M:E ratio increases
 Anemia: 3:3  1:1  M:E ratio decreases C. ANEMIA WITH VITAMIN B12 OR FOLATE
DEFICIENCY
5. OTHER SPECIAL TEST DIAGNOSTIC OF THE  Nutritional deficiency anemia: IDA, B12 or
SPECIFIC TYPE OF ANEMIA folate deficiency anemia
 AIHA: serologic test: antibody testing to
indentify the specific autoantibody D. ANEMIA ASSOCIATED WITH IMPAIRED BONE
 Pernicious anemia: Schilling’s test MARROW OR STEM CELL FUNCTION
 Aplastic anemia; bone marrow is infiltrated with
CLASSIFICATION OF ANEMIA fats thus reduced or no red marrow that is the
- Some anemia have more than 1 pathologic
active marrow producing blood cells.
mechanism (e.g. B12 deficiency  tapeworm
infection + nutritional deficiency + antibody) and
E. ANEMIAS ASSOCIATED WUTH DECREASED RED
go through more than one morphologic stage
CELL SIRVIVAL AND INCREASED RED CELL
(IDA: microcytic-hypochromic; normocytic-
DESTRUCTION
hypochromic)
 Prone to damage: hemolytic anemia
1. MORPHOLOGIC CLASSIFICATION
2. ETIOLOGIC CLASSIFICATION
F. ANEMIA SECONDARY TO BLOOD LOSS
3. PHYSIOLOGIC CLASSIFICATION
 Hemorrhagic anemia:
 The ability of the bone marrow to respond
o Chronic blood loss
physiologically by producing erythrocytes is
o Acute blood loss iii. Post-acute
measured by RPI  Ineffective
hemorrhagic anemia  IDA
erythropoiesis (RPI <2.0)
 Effective erythropoiesis (RPI >3.0)  e.g. RPI= 4
means that the rate of production has increased
4x than its usual for compensation

NOTE:
Some anemia have more than 1 pathologic
mechanism & go through more than one morphologic
stage.

ETIOLOGIC CLASSIFICATION OF ANEMIAS: CAUSE OF


ANEMIA
A. RELATIVE ANEMIA
 Increase in plasma volume  hemodilution
 RBCs are NOT reduced but is diluted with
increase plasma volume
 TRUE/ABSOLUTE ANEMIA: reduced in number
of RBC due to defective production
HEMATOLOGY 2

MORPHOLOGIC CLASSIFICATIONS OF ANEMIA


- Usually followed
- Basis: size and hemoglobin

MCV: according to size MCHC: hemoglobin concentration RDW: a measure of the degree of
<80 fL = microcytic MCH is not helpful in morphological anisocytosis and poikilocytosis  used
80-100 fL = normocytic classification of anemia because it always because MCV and MCHC cannot be rely
>100 fL = macrocytic follow the MCHC upon when the cell population is diverse.
<32% = hypochromic
32-36% = normochromic machine gives its value from histogram
>36% = spherocytic
According to MCV and MCHC: single
population
3 CLASSIFICATION OF ANEMIA
1. Normocytic – normochromic
2. Microcytic – hypochromic
3. Macrocytic – normochromic
4. Normocytic - hypochromic  gradual
changes seen in IDA

MCV: Single red cell index that can be used to CBC  anemia  retic count  morphologic classification
morphologically classify anemia; will suffice alone 
microcytic; macrocytic; normocytic

MICROCYTIC: the most common anemias included are


those
characterized by defect in hb: heme: SDA, IDA; globin: ACI,
Thalassemia  in general thalassemia fall under
microcytic anemia

GLOBIN DEFECTS: deficiency in alpha and beta chain


synthesis: thalassemia; defect in amino acid
structure/composition: hemoglobinopathies  fall under
normocytic anemia (Hb C and S) except Hb E disease or
trait.
HEMATOLOGY 2

MACROCYTIC: include problems in DNA synthesis 


megaloblastic anemia

NORMOCYTIC: more on destruction (hemolytic) and


decrease in production (aplastic: BM failure)

MICROCYTIC HYPOCHROMIC MACROCYTIC HYPOCHROMIC NORMOCYTIC NORMOCHROMIC


Main cause: defective hb synthesis Commonly Macrocytic Commonly Normocytic
Heme synthesis: IDA, SDA, ACI Globin  Vitamin B12 deficiency  Hypoproliferative anemia
synthesis: alpha or beta thalassemia and anemia  Folic acid  Myelophthisic anemia
Hb E disease deficiency anemia  Hemolytic anemia
 Hemoglobinopathies
Commonly Microcytic Occasionally Macrocytic  Acute blood loss anemia
 Iron deficiency anemia  Hypoproliferative anemia  Anemia of chronic disease
 Sideroblastic anemia   Refractory anemia  Renal and endocrine
Thalassemia  Liver disease diseases
 Hemolytic anemia  Paroxysmal nocturnal
Occasionally Microcytic  Acute blood loss anemia hemoglobinuria
 Anemia of chronic
disease/inflammation Occasionally Normocytic
 Hemoglobinophaties (Hb E  Early iron deficiency
disease  Refractory anemia
The cells will keep on dividing until their optimum MCHC is
reached. In normal erythropoiesis, it takes only 4-5
divisions for mitosis  Pronormoblast will divide once
producing 2 basophilic normoblast (BN) and these BN will
divide twice and the last to go division is the rubicyte  4x
division = 16 ; 5x division = 32

Every time the cell divides the cell size becomes smaller

In microcytic-hypochromic, since due to decrease hb, it


will not reach its optimum MCHC therefore it will keeps on
dividing thus decrease in cell size Microcytic: extra division
Hypochromic: decrease Hb.
HEMATOLOGY 2

MICROCYTIC- HYPOCHROMIC ANEMIAS intestine (particularly in


 ETIOLOGY: Microcytosis is caused by one or duodenum and jejunum) they
more extra cell division due to the depression of will be absorbed by mucosal
the MCHC. This conditions also exhibits cells and immediately cause
hypochromia which is cause by an impairment in oxidation of ferrous thus
the hemoglobin synthesis due to abnormalities converting back into ferric form.
in either the heme or globin synthesis or both. The ferric form will be released
into the plasma circulation by
1. IRON DEFICIENCY ANEMIA (IDA) the mucosal cells.
 Iron Metabolism: Iron is absorbed in the ferrous o Fe+++  bloodstream
form, later oxidized in the mucosal cells to the  Fe binds with apoferritin to form
ferric form. Absorbed iron passed on directly to FERRITIN (major storage form of
the blood stream by the mucosal cells, but most iron  stored in tissues and
of it is bound to Apoferritin, which then forms liver).
Ferritin. Iron in the bloodstream is transported  Fe-transferrin  transported to
by Transferrin which delivers it to the Mononuclear Phagocytic Cell
mononuclear phagocytic cells (MPC) of the (MPC) going into the bone
bone marrow and other issues. During marrow and liver.
erythropoiesis, the MPC of the bone marrow
NOTE:
incorporated iron into the developing red cell.
 In bone marrow, during erythropoiesis, when
 NORMAL BODY IRON (FE): 4,000 mg
the developing red cell start to synthesize hb,
o 3 COMPARTMENTS: iron is distributed mononuclear cell will transport and
among these compartments and sub- incorporate iron into developing RBC 
compartments. incorporation is called ROPHEOCYTOSIS
 STORAGE: in the form of ferritin  ROPHEOCYTOSIS: During hemoglobin
(major form in the liver and synthesis in immature red cell, mononuclear
tissue) and hemosiderin cell will incorporate the iron thus iron (Fe+++)
 TRANSPORT: protein will be released and immediately reduced into
transporter for iron ferrous (Fe++) and incorporate itself into
 FUNCTIONAL: hemoglobin, protoporphyrin IX to form heme.
myoglobin, iron-containing
enzyme. Most of the iron is in
the hemoglobin approximately
60%; thus deficiency in iron will
affect hemoglobin resulting in
anemia.
 IRON KINETICS:
o Fe++ & Fe+++ (diet)  Fe++ (stomach)
 Fe+++ (Mucosal cells)
 What the body can absorb is
only the ferrous form; the ferric
form can be reduced into
ferrous form in the stomach by
our gastric juices (very acidic
due to HCl) to promote ETIOLOGY/ RELATED CAUSES: IDA
absorption, when the ferrous 1. CHRONIC BLOOD LOSS
form of iron reaches the small  Most common cause of IDA in adults.
HEMATOLOGY 2

 Small volume but chronic Solubilizing agents: Precipitating agents:


 Male: GIT bleeding  unnoticed because it sugars and amino acids phosphates
occurs inside unless it develops into other Iron deficiency Iron excess
serious complications. Increased erythropoiesis Decreased erythropoiesis
 Female: prolonged menstruation, bleeding infection
after childbirth or cesarean
 ACUTE BLOOD LOSS will NOT lead to IDA
STAGES IN THE DEVELOPMENT OF IDA
2. INCREASE DEMAND  STAGE I: Iron depletion stage: Iron is mobilized
 especially among pregnant women (30- from stores, storage iron decreases, plasma
75mg), adolescents and infants
ferritin decreases, iron absorption increases.
 Not pregnant female: 10-20 mg thus female
o First to decrease is iron storage
needs more iron due to monthly blood loss
compartment which is measured by
 Pregnant: ferrous sulfate supplement to
prevent IDA transferrin13
 Adolescent: fast growing  more iron needed o Serum ferritin first decreases and
by the body reflects the iron storage or ferritin
 Infants: breastmilk – poor in iron  pure o Heme is consisting of iron and
breastfed infants may develop IDA called protoporphyrin IX. Deficiency in iron
“milk anemia of infancy”  Male has less iron would lead to increase in
demand because they do not have monthly protoporphyrin IX and will accumulate
menstruation/blood loss inside the RBC which is measured as Free
Erythrocyte Porphyrin  FEP is increase
3. MALABSORPTION o Since functional iron is still normal then
 Absorption of iron happen in small intestine hemoglobin and hematocrit are normal
(duodenum and jejunum). thus, it is characterized as normocytic
 Regardless of the amount of iron take in, the and normochromic
absorption is limited to 10% of dietary iron
o Normal serum iron and TIBC
 Celiac disease (sprue): disorder characterized
 STAGE II: Iron deficient erythropoiesis: after
by antibody destruction of the small intestines
 destroyed absorption sites  total iron stores are depleted, the plasma iron
gastrectomy: decreased absorption sites concentration falls, saturation of transferrin falls
 Intestinal Parasites: below 15% and the percentage of sideroblasts
o N. americanus: 0.03 ml/day  per decreases in the marrow.
worm o Iron storage (ferritin) and transport iron
o A. duodenale: 0.15 – 0.25 ml/day  is depleted
per worm o Decrease in serum iron with increase
o T. trichiura: 0.005 ml/day  per FEP
worm o Increase TIBC: indirect measurement of
transferrin
4. POOR DIET o Transferrin: transport two molecules of
5. FREQUENT BLOOD DONATION (225 gm/unit)
iron simultaneously
 450 ml: blood donated
 When transferrin is bound into
iron because iron is normal, it is
PROMOTE ABSORPTION REDUCE ABSORPTION fully saturated thus it will no
Ferrous form Ferric form longer bind with additional iron.
Inorganic form Organic form In the absence of iron (IDA),
Acids: HCl, Vitamin C Alkali: Antacids transferrin remains to be free.
Pancreatic secretions
HEMATOLOGY 2

When added with the reagent - In early IDA, RBCs are N/N; in later stages, RBCs
iron, it will bind with iron. become Microcytic-Hypochromic. Severe cases
 Transferrin when it is not will exhibit poikilocytosis & anisocytosis 
saturated, its binding capacity Reticulocyte count is low; Platelet count is
for iron is increased measured slightly elevated
by TIBC. - Bone marrow smear is negative for hemosiderin
o Functional iron is still normal thus the granules
anemia is normocytic – normochromic - Serum iron is decreased, and serum ferritin may
 When the cells are in the be reduced to zero; increase Free Erythrocyte
circulation, they are fully Protoporphyrin (FEP); increase Total Iron-
hemoglobinized and in the Binding Capacity (TIBC)
presence of IDA, the mature RBC - General blood picture: microcytic-hypochromic
will not be affected because the with poikilocytosis and anisocytosis and elevated
mature RBC have fully RDW.
synthesized their hemoglobin. o RDW: measure in the variation of size
 IDA affects the developing RBC and shape
in the bone marrow wherein the - Reticulocytopenia: Even though the red marrow
bone marrow will immediately tries to compensate, the red cells are
suffer in decreased in iron ineffectively growing therefore
because of the storage form of reticulocytopenia.
iron. - Slight thrombocytosis: If it is associated with
 Aside from the liver and tissues bleeding because bleeding is also one of the
having the storage form, there is causes of IDA, chronic bleeding (most common
also in the bone marrow which cause of IDA) especially in adults, it may also be
will be first depleted. characterized by slight thrombosis.
 STAGE III: Iron deficiency anemia: anemia is now - Prussian blue staining: hemosiderin granules are
detectable. The anemia is at first normochromic negative.
acid normocytic, gradually becomes microcytic
IRON PROFILE TESTS FOR THE DIFFERENTIAL DIAGNOSIS
and finally microcytic & hypochromic.
OF IRON DEFICIENCY ANEMIA
o Iron deficient red cells from the bone
marrow will be released and will appear  SERUM FERRITIN
as microcytic as reflected by the low o Reflects the body’s tissue iron stores
hemoglobin (hypochromic). o Decrease in iron causes increase
o Findings: decrease hemoglobin, serum protoporphyrin 9. Though normally, RBC
iron, ferritin ; increase TIBC produces more protoporphyrin than
iron, but in the absence of iron, the more
the protoporphyrin will accumulate
causing increase FEP. It is free because it
is supposed to bind with iron but since
there is no iron, it remains to be free
erythrocyte protoporphyrin. Some
measured it as ZEP because the FEP can
combine with zinc and measured as zinc
erythrocyte protoporphyrin
o Reference value: 12-300 ug/dL
LABORATORY FINDINGS OF IDA
 SERUM IRON
HEMATOLOGY 2

o A sensitive indicator or iron depletion CLINICAL FEATURES OF IDA


but has a marked diurnal variation by as
 TONGUE- ATROPHY: The tongue is red because
much as 30% with its highest values in
of iron in heme present in hemoglobin in blood
the morning and lowest values late in
and myoglobin in tissues, but since it lacks iron,
the day.
it will atrophy (become smaller).
o Reference value: 50-160 ug/dL
o Decreased in IDA  ANGULAR STOMATITIS: the lips will get dry and
have cracks
 FREE ERYTHROCYTE PROTOPORPHYRIN (FEP)  CHEILITIS: the drying and inflammation of the
o Normally, red cells produce slightly more lips; associated with IDA
protoporphyrin than what is needed, but  “Webs” of tissue or partial strictures at the
when iron is deficient, protoporphyrin junction of the esophagus and hypopharynx:
build up in the erythrocytes there will be cracks and wounds inside 
o Reference value: 10-99 ug/dL resulting now to difficulty in swallowing; affects
o Increased in IDA; decreased in most entire GIT.
SDAs.  CHRONIC GASTRITIS – decreased gastric fluid
secretion which is important because it is the
 TOTAL IRON BINDING CAPACITY (TIBC) gastric juice that is acidic which helps in the
o Binding capacity: measure the ability of absorption of iron. The acidity of gastric juices
transferrin to bind with iron. Wherein, if helps reduce the ferric into ferrous in order to
tranferrin is not bound to iron, it will absorb the dietary iron even in the ferric form.
bind. But if the transferrin is already  NUMBNESS & TINGLING SENSATION: peripheral
saturated with iron, it can not further tissues nerves will be affected especially the
bind with other iron (in the reagent). distal parts of the body (toes, fingers, and feet).
o Wherein in normal iron status, iron is  KOILONYCHIA: distortion of the nail bed, spoon
plenty thus transferrin carry two iron shaped of the nails due to lack of iron in the
molecules. tissues
o In IDA, iron is depleted thus they carry  PICA: unusual and compulsive appetite for a
no iron thus transferring is free. When certain kind of food or non-food item; among
the reagent (iron) is added, the children with severe IDA wherein they even have
transferrin can bind with it because TIBC abnormal craving for unusual food.
is increase.  SPLENOMEGALY: spleen function to filter blood
o Reference value: 250-400 ug/dL and remove a lot of abnormal cells so it will be
o Increased in IDA overworked and abnormal cells will clog in the
splenic circulation resulting enlargement of
 TRANFERRIN SATURATION/ PERCENT spleen.
SATURATION OF TIBC
NOTE:
o Measures how transferrin is saturated; Aside from the general manifestations of anemia,
percent saturation other manifestations include paresthesia, pica,
o Reference value: 20-55% koilonychia, atrophy of epithelium of the tongue with
o Decreased in IDA burning or soreness, fissures/cracks or ulcers at the
corners of the mouth, difficulty swallowing owing to
NOTE:
webs of tissue or partial strictures at the junction of
Since iron is not only for hemoglobin synthesis but also the esophagus & hypopharynx. Chronic IDA also
important for the tissues, enzymes, myoglobin and manifests gastritis and splenomegaly.
other proteins, these tissues and proteins will also be
GENERAL BLOOD PICTURE OF IDA
damaged due to IDA.
 Microcytic-hypochromic
HEMATOLOGY 2

o Central pallor: >1/3 diameter  SDA are a group of heterogeneous disorders


o Normal RBC has the same size as the characterized by hypochromic red cells, which
small lymphocyte are microcytic in the hereditary forms and often
 So roughly, even with the absence of MCHC macrocytic in the acquired forms, as a result of
computation; blood picture alone can suffice defective heme synthesis, the red cells may also
that the cells are hypochromic  central be mixed with normochromic cells (dimorphic
pallor is greater than 1/3 of the diameter.
appearance).
 Associated with defective protoporphyrin
TREATMENT OF IDA synthesis which leads to excessive accumulation
of iron. The unused iron is deposited in the
 Dependent on the cause of IDA. mitochondria of normoblasts as siderotic
 Important to consider the etiology for the granules.
treatment of anemia.
 Since iron is primarily deficient, iron supplement NOTE:
is given. In SDA, the first enzyme (D-ALA synthase) is commonly
 If the main cause of IDA is malnutrition, ferrous affected or if not the last enzyme (ferrochelatase). If
sulfate supplement may be given. the D-ALA synthetase is affected, defective or
deficient, it will slow down the very first reaction
 If the cause is chronic blood loss, blood loss must
which is the condensation between succinyl CoA and
be supervised and give iron supplement to glycine and deficiency in this reaction would lead to
monitor the progression of disease. deficient D-ALA and the following reactions will
 If it is caused by parasite, eradicate the parasite. become retarded or slow down thus protoporphyrin
IX will also be deficient. When iron is transported in
NOTE: RECAP
the RBC, it will not be utilized because of the
IRON DEFICIENCY ANEMIA
deficiency in protoporphyrin IX; wherein iron is
 Described as microcytic: hypochromic type of supposed to bind with protoporphyrin IX but in the
anemia absence of protoporphyrin IX, the iron will be
 Morphologic changes: At first it is normocytic unutilized and the non-utilized iron will precipitate
and as it becomes more severe, it becomes inside the RBC, inside the mitochondria of RBC
microcytic. becoming now the siderotic granules. When siderotic
granules are found surrounding the nucleus of RBC, it
IRON DISTRIBUTION: 3 POOLS/ COMPARTMENTS is called the ringed-sideroblast which are immature
a. IRON STORAGE POOL: includes ferritin RBC containing siderotic granules which are non-
measured using serum ferritin and utilized iron.
hemosiderin measured via Prussian blue
staining.
b. TRANSPORT COMPARTMENT: iron bound to
transferrin measured as serum iron or
transferrin saturation or total iron binding
capacity (TIBC).
c. FUNCTIONAL IRON COMPARTMENT: iron
present in functional proteins such as
hemoglobin, myoglobin and other enzymes.

2. SIDEROBLASTIC ANEMIA (SDA)


 It is a group of anemias characterized by
COMPARISON:
abnormalities in the synthesis or enzyme
 In IDA, iron is deficient thus increase in
involved in synthesis of heme. protoporphyrin IX
o Negative in prussian blue staining
HEMATOLOGY 2

 In SDA, protoporphyrin IX is deficient thus iron B. AQUIRED SIDEROBLASTIC ANEMIA


will not be utilized and precipitate as siderotic  Due to exposure
granules  Associated with macrocytosis
o Not in all types of SDA, the  Acquired Idiopathic Sideroblastic Anemia
protoporphyrin IX will be decrease. (AISA)/ Refractory Anemia with Ringed
There are some types wherein
Sideroblast (RARS)
protoporphyrin IX will be increased.
o Mostly encountered in elderly patients
o Dimorphism is observed in
sideroblastic anemia. (older than 50 y/o) affecting both sexes.
o Some cases, protoporphyrin IX is Actual etiology of the disease is still
increase or decrease depending on unknown but consistent findings
which stage in the heme synthesis is included decreased activity of ALA-
inhibited. synthetase or an abnormal high
o SDA is highly positive in Prussian blue requirement for the cofactor pyridoxial-
stain since iron is not utilized 5PO4 to maintain ALA synthetase RBCs
are hypochromic, despite presence of
large quantities of iron.
CLASSIFICATION/ TYPES OF SDA
 Secondary Sideroblastic Anemia: Secondary to
1. INHERITED some agents that interfere with heme synthesis
 Caused by an abnormal gene (Lead Poisoning; Alcohol, Drug-induced)
 Inability to inherit the gene needed by the o Known etiology
enzyme
 Characterized by microcytic cells

2. ACQUIRED
OTHER RELATED DISORDER:
 Exposure to agent that will inhibit the function of
the enzyme like in lead poisoning wherein lead  PORPHYRIAS:
interferes with some of the enzymes involved in o Group of disorders characterized by the
heme synthesis. accumulation of porphyrins.
 Characterized by macrocytic cells. o Diseases characterized by impaired
 Example: lead poisoning  lead may interfere in production of heme (can be hereditary
the synthesis by inhibiting pyridoxal phosphate or acquired) causing porphyrins to not
ALA synthetase, ALA dehydrase or be completely changed and metabolized
ferrochelatase. When ferrochelatase or heme resulting them to accumulate inside the
synthase (which catalyzes the addition of RBC or liver cells.
protoporphyrin IX with iron) is inhibited by lead o The causes of porphyria can be the same
 protoporphyrin IX may no longer bind with with those of SDA and the more
iron thus there would be protoporphyrin IX and prominent feature is the accumulation
iron build up of porphyrins.
 Lead poisoning can also lead to
porphyrias
A. HEREDITARY SIDEROBLASTIC ANEMIA o Acquired types are usually associated
 Sex-linked recessive trait, mostly caused by with enzyme deficiency. The products
decreased in D-ALA synthase activity from earlier stages (porphyrins) in the
 Deficiency of D-ALA synthase causes the pathway accumulate in the cells that
retardation of heme synthesis pathway actively produce heme proteins such as
 Associated with microcytosis RBC and hepatocytes. These porphyrins
HEMATOLOGY 2

can also be excreted in the urine or feces  Most common clinical features:
or be deposited in body tissues. Photosensitivity and Psychiatric or neurologic
symptom due to deposition of porphyrins
NOTE: PORPHYRIA  Hematologic manifestations: Acute
MAIN FEATURE: accumulation of porphyrins intermittent porphyria (abdominal pain),
 Porphyrins accumulate first in the RBC congenital erythropoietic porphyria (port
because RBC is involved in heme synthesis and wine red urine), variate porphyria (psychosis
in the liver because it is involved in the and photosensitivity) & erythropoietic
synthesis of myoglobin. protoporphyria (psychosis)
o During damage of RBC, when it is
hemolyze, as it reaches its 120th day,
the RBC will hemolyze and all its
contents will diffuse out into the
plasma including the porphyrins.
o When liver cells are damaged, they
also release their contents like,
porphyrins into the plasma.
Porphyrins will be elevated in the
plasma and can be filtered by the
kidneys to appear in the urine HEMOCHROMATOSIS
(porphyria) where the urine appears
 Another disease related to Iron metabolism:
as port wine red. Some are excreted
in the feces. TRUE IRON OVERLOAD.
o It is more accurate and precise to  TO COMPARE: SDA  Iron wasn’t used in the
measure porphyrins in the feces than phase of normal iron (NORMAL: Iron; Cannot be
in urine. used/metabolized = NO IRON OVERLOAD)
o Deposit in body tissues – when  NORMAL: RATE of IRON ACQUISITION (1%) =
porphyrins deposit in the skin, the RATE of IRON LOSS (at least 1% of Iron daily = Red
skin becomes photosensitive. cell destruction 1% daily)
Porphyrins absorbs the zorret band o Rate of Loss (1%) daily is replaced by
coming from the sun, so when what is absorbed (1%) daily
exposed to sun, the skin of the patient
 HEMOCHROMATOSIS: RATE of IRON
suffers from sun burn, blindness
ACQUISITION > RATE of IRON LOSS
(eyes), brain damage or psychiatric
abnormality (brain). o Iron acquisition exceeds the rate of iron
o Some teeth and bones especially loss  iron is accumulated in the body
among the growing children wherein  Results when the body’s state of iron acquisition
some of the substances will exceeds the rate of iron loss. It can be acquired
fluorescence. Teeth and bones are or hereditary (mutations affecting the proteins
fluorescent because of the deposited of iron metabolism). The body’s first reaction is
porphyrins. to store excess iron in the form of ferritin, then
in the form of hemosiderin within cells.
PORPHYRIA: associated with vampire stories
 A common disease affecting the royal families CAUSES OF HEMOCHROMATOSIS
in Romania and Transylvania.
 ACQUIRED: Transfusion-related
 Characterized by bleeding teeth due to lack of
hemoglobin; evasion from sunlight because o It cannot be due to DIETARY REASONS,
they are prone to suffer sunburns and excessive iron in the diet is unlikely to
blindness; fluorescing teeth due to deposition cause hemochromatosis because our
of fluorescing porphyrins in the bone absorption for Iron is LIMITED
HEMATOLOGY 2

o Commonly caused by TRANSFUSION- o Fe++ (+O2)  Superoxide and Free


RELATED radicals – strong oxidizing agents 
 IDA: one factor leading to this PEROXIDATION of membrane lipids
condition is Frequent Blood results to:
Donation that every time we  Cell respiration - (Destroying
donate blood we lose iron. Mitochondrial Membrane) –
 Hemochromatosis: Every time Impaired ATP production = No
the recipient receives blood Cellular Respiration
(transfusion) the recipient gains  Lysosomal enzymes - (Impairing
Iron. Lysosomal Membrane) -
• Example: Patients with Beta- Rupture = Release of Hydrolytic
Thalassemia would require Enzyme
frequent donation or  Lysosome = Suicide Bag of the
transfusion of blood (PRBC/WB) Cell because once it releases its
they gain Iron thus repeated hydrolytic enzymes it can lead to
transfusion will lead to Autolysis (Cell will digest itself)
Transfusion-Related cell damage it will be destroying
Hemochromatosis. the nucleus, nuclear membrane
and in general will cause an
 INHERITED: Mutations affecting the proteins of irreversible membrane damage
iron metabolism of the cell and the organelles.
o Results to Less Metabolism: More Iron  Irreversible membrane damage
Accumulation. (Cell Membrane)
o Excessive Iron (Iron Overload): most of o In the presence of Oxygen, free ferrous
the Iron will first be stored as ferritin, but iron initiates the generation of
when overwhelmed will be stored as superoxide and other free radicals,
hemosiderin. which results in the peroxidation of the
o 2 Storage form of iron: 1st - Ferritin  membrane lipids.
when overwhelmed  2nd - o Results to damage of cell membrane,
Hemosiderin nuclear membrane, mitochrondrial and
o Both storage forms have limits and when lysosomal mnembranes. These events
both get overwhelmed, the remaining ultimately affect cellular respiration,
excessive Iron will have nowhere to be enzyme digestion, cell death, and organ
stored thus the remaining Iron (Fe++) in damage.
the presence of free oxygen (O2) will o Hemosiderin also deposits into tissues
now cause the generation of and organs leading to further organ
SUPEROXIDES and FREE RADICAL which damage:
are TOXINS and STRONG OXIDIZING
AGENT that cause oxidation of proteins,
lipids and cells  causing
PEROXIDATION of Membrane Lipids,
Cell Membrane, Mitochondrial
Membrane, Lysosomal Membrane and o SKIN AND PANCREAS: “bronze diabetes” –
other organelles, even the Nuclear bronze skin plus diabetes meaning the
Membrane leading now to impaired ATP Hemochromatosis have already affected the skin
production. and pancreas.
HEMATOLOGY 2

 SKIN: Bronze/ golden color due to o Free Oxygen Reaction


cellular damage and accumulation of o But itself can be overwhelmed and will
excessive iron. The skin will absorb the only be able to SLOW DOWN the CHAIN
excessive Iron and it will accumulate of REACTIONS = Inefficient
resulting to a BRONZE SKIN COLOR o It should be alongside the
 PANCREAS: Pancreas will be destroyed Therapeutic/repeated phlebotomy and
and its production of Hormones Iron-chelating drugs
(regulate Carbohydrate Digestion) like
NOTE:
INSULIN, GLUCAGON etc.  with that it
BOTH IDA and SDA  heme synthesis defect
affects the Glucose Metabolism and can
Thalassemia  globin synthesis defect
lead to Diabetic Mellitus
 Hemoglobin are usually dimers, their globin
o LIVER: Cirrhosis-induced jaundice subsequent portion has 4 polypeptide chains  2 different
Liver Cancer types of chain (alpha and beta).
o HEART: Congestive Heart Failure o Example: A1 (heme + globin)  4 PPC
o Other more organs can be passively destroyed (2 alpha and 2 beta chains)
o To synthesize the chains, it need
SCREENING TEST FOR HEMOCHROMATOSIS:
genes (complete number) that is
 Transferrin saturation (common test performed) inherited from parents (one per each
and Ferritin (excessively overwhelm). parent).
 Alpha chain: needs 4 alpha
 Measurement of Transferrin Saturation
genes
o Transferrin is fully saturated due to
 Beta chain: 2 beta gene
excess Iron/ Iron overload ELEVATED o Each PPC has its amino acids
Transferrin Saturation  Alpha chain: made up of 141
 Transferrin and Iron-binding Capacity(TIBC) will AA  per chain
be DECREASED  Beta chain: 146 AA  per
o NOT THE APPROPRIATE TEST for chain
HEMACHOMATOSIS  HEMOGLOBINOPATHIES: condition
characterized by qualitative structural
TREATMENT: abnormalities of the globin polypeptide chains
 THERAPEUTIC/REPEATED PHLEBOTOMY that result from alteration of the DNA genetic
code for those chains.
o Most practical treatment to prevent
o Structural defect due to defect in
excessive Iron and prevent the
amino acids; classified as normocytic
successive chain reaction anemia except Hb E disease
o Some patients require 2-3 times removal  THALASSEMIA: diverse group of genetic
of blood/phlebotomy  removal of disorders characterized by a primary,
excessive Iron quantitative reduction in globin chain
 IRON-CHELATING DRUGS to bind/chelate excess synthesis for hemoglobin
iron for safe excretion. o Synthesis defect (alpha or beta) 
o Desferrioxamine/Desferal disorder characterized by defective or
 Intake of Vit. E and C deficient number in chains due to
o Slow down chain of reaction but effect deletion of some gene thus proteins
will be overwhelming  inefficient; that is encoded by the gene can’t be
synthesize.
retard.
o Can somehow help because they are a
REDUCING SUBSTANCE (especially Vit. E)
resulting to the RETARDATION of the
Iron and
HEMATOLOGY 2

3. THALASSEMIAS CLASSIFICATION CLINICAL HEMOGLOBI


 Thalassemias are the results of impaired MANIFESTATIO N
(deficient) globin chain synthesis. These may N
affect either the alpha or beta chain synthesis. Silent carrier Normal 3 functional
(one-gene hematologically, alpha gene
Greek No. of No. of Chromosome deletion) with few target A1= normal
Name genes Amino Location cells, elliptocyte (97%)
Acids
Alpha 4 (2 141 16 s/s:
mother; 2 asymptomatic
father)  normal
Beta 2 (1 146 11
mother; 1
father) Alpha- Mild anemia, 2 functional
Delta 2 146 11 Thalassemia trait: microcytic- alpha gene
Gamma 2 146 11 (two-gene hypochromic, remaining
Epsilon 2 146 11 deletion) elliptocytes, thus
Zeta 4 146 16 target cells synthesis of
(Rodak’s) Hb A1 is
slightly
decresed:
normal A1
levels atleast
97%.

Hb A (60%);
Hb Barts
(510%)
A. ALPHA THALASSEMIA HbH disease Microcytic, One
(Three-gene hypochromic, Hb functional
o A deficiency in the synthesis of Alpha-
deletion) H inclusions, gene
globin chains. Each individual has two
high retics remaining
sets of two alpha genes. Suppression of that
all four genes is needed to completely A1: 2 alpha & 2 is not enough
suppress alpha chain synthesis. The beta to cause
usual mechanism of suppression is by A2: 2 alpha and 2 normal
gene deletion. delta production of
o Main common cause: gene deletion or Hb F: 2 alpha and alpha
no inherited gene. 2 gamma polypeptide
o Deletion is indicated by blank: e.g. – Since alpha chain.
a/aa  one gene deletion; --/aa  two chain production
deletion; --/a  three deletion; --/--  is affected Hb H  seen
and these an pitted golf
four gene deletion.
hemoglobin ball in PBS.
needs the alpha
chain thus their
production will
also be
HEMATOLOGY 2

retarded  RBC CLASSIFICATION OF BETA THALASSEMIA


will compensate Beta Beta Beta
by Thalassemia Thalassemi Thalassemi
increasing the Major/ a Major/ a Major/
synthesis of beta Cooley’s Cooley’s Cooley’s
chain  anemia anemia anemia
4 beta chain 0 0 + +
(B l B ) (B l B ) (B0l B)
producing Hb H
•Complete lack of •Caused by a •Sufficient
Bart’s Hydrops Stillborn, Hb Bart’s:
Beta globin partial quantities of
fetalis (four-gene hydrops tetramer Hb
production suppression of HbA1 is
deletion) (enlarged head) consisting of
•Most affected the Beta genes produced.
-Hb Bart’s has 4 gamma
patients exhibit •Symptoms •Hemoglobin
abnormal chains.
retarded growth are similar levels are
function
with mongoloid with that of slightly low,
wherein it can’t Since there is
facial features and Beta but blood
promote no
severe anemia Thalassemia smear shows
oxygenation of production of
•Marked by major, but similar
tissues. alpha chains
characteristics depend on the morphology as
due to total
changes in RBC extent of gene in Thalassemia
deletion of
morphology; suppression Major or
alpha gene,
microcytosis, • May produce Intermedia
the RBC will
hypochromia, varying •HbF is about
compensate
anisocytosis and amounts of A1 2-3%
by producing
poikilocytosis. depending on •HbA2 level is
gamma
Numerous target the degree of elevated 
chains thus
cells, Howell-Jolly suppression has delta chain
formation of
bodies and
Hb bart’s.
siderocytes. TYPES:
•HbF level is Homozygous
elevated (40-60%) (B+l B+): 2 beta
• Can synthesis A2 gene is
• Most severe of partially
all B-thalassemia suppressed
that requires Heterozygous
B. BETA THALASSEMIA frequent (B+IB): One
o Characterized by a deficiency in Beta- transfusion gene is
globin chain synthesis. Each individual partially
has one set of two beta genes. suppressed
Suppression or absence of these beta and the other
one is normal
genes results to deficient beta chain
synthesis.
o Gene deletion is indicated by 0
HEMATOLOGY 2

NOTE:
Hereditary Persistence of Fetal Hemoglobin and B-
Thalassemia Major: highest concentration of Hb F can
be seen.

1. Altered Iron Metabolism  Microcytic


 Among the 4 pathophysiology, most common to
happen due to increase in acute-phase protein
In thalassemias and other disorders characterized by and it is a direct effect of inflammation
abnormal hemoglobin synthesis like IDA and SDA, it is
very common to see target cells  nonspecific finding 2. Direct inhibition hematopoiesis, decreases
unlike schistocytes that is a hallmark finding in production of EPO. moderate destruction 
hemolytic anemia indicating hemolysis and teardrop normocytic (do not involve iron)
cells in myelofibrosis
 During acute/chronic inflammation, the
macrophages and lymphocytes are activated
thus produce cytokines of different types that
includes tumor necrosis factor-alpha (TNF-
alpha), Interferon-gamma and other
interleukins. Some of these cytokines will cause
inhibition of bone marrow function leading to
4. ANEMIA OF CHRONIC INFLAMMATION (ACI) decrease hematopoiesis, inhibition of kidney
 These diseases are grouped into chronic function thus decreased erythropoietin
inflammatory diseases and malignant diseases. production and some may cause direct damage
It is the inflammation that may lead to anemia. on RBCs
 Malignant disease/ cancer NOTE:
 Chronic inflammatory disease Anemias that develop in chronic diseases are caused
o Infectious TB by the effects of the increased inflammatory
o Osteomyelitis cytokines. (i.e. interleukins) produced by activated
o Non-infectious RA cells during the chronic inflammation: These cytokines
o SLE and Crohn’s disease may cause any of the following mechanisms:
1. ALTERED IRON METABOLISM OR IMPAIRED
MECHANISMS OF ANEMIA OF CHRONIC DISEASES: FERROKINETICS
 Activated lymphocyte will produce IL-6 that
will act on the liver, it will activates the liver to
produce increase hepcidin (Positive Acute
Phase
 Protein: proteins that immediately increase
during inflammation) caused by an increase of
the following acute phase reactants that
interfere with iron metabolism:
o HEPCIDIN: this causes the
degradation of the transmembrane
protein ferroportin used by
macrophages and hepatocytes to
export iron into plasma.
HEMATOLOGY 2

 Increase in hepcidin will result o Hb C (B6 glu  lys) and Hb S(B6 glu 
to failure of iron to be val) has structural defect but Hb E has
released in the plasma both structural and synthesis defect
because iron will be retain in  Decrease in B-chain synthesis
the cells  2nd most common hemoglobinopathies
 Decrease iron absorption and
release  decrease plasma
iron
 FERROPORTIN: transport
protein found in the intestinal
enterocytes(cells of
duodenum), hepatocytes,
and macrophages
o LACTOFERRIN: An iron-binding
protein in the tertiary granules of MACROCYTIC ANEMIA
neutrophils. Its avidity for iron is  MCV: >100 fL
greater than that of transferrin.  A macrocytic cell seems to have more
During inflammation, it scavenges hemoglobin content compared to normocytic
available iron at the expense of but in relation to its size it is normal. 
transferrin. Macrocytic  normochromic.
o FERRITIN: developing RBCs do not
have a ferritin receptor, thus, cannot TWO TYPES OF MACROCYTIC ANEMIA:
utilize iron for hemoglobin synthesis.
 Increase in the production of  In cell maturation, the nuclear-cytoplasmic
apoferritin that binds with maturation must be synchronous in order for the
iron to form ferritin, thus iron cells to be normocytic when released. In
will not be utilized because it asynchronous maturation, the maturation of
is trap in ferritin. nucleus and cytoplasm is not the same that can
2. MODERATE INCREASE IN THE DESTRUCTION lead to megaloblastic maturation.
OF RBC
3. DECREASED PRODUCTION OF 1. NON-MEGALOBLASTIC
ERYTHOPOIETIN  Normal maturation therefore normal
 Production of inflammatory cytokines (such as development however when released in the
tissue necrosis factor-a and interleukin-1 from
circulation they appear large
activated macrophages and interferon-g from
 PATHOPHYSIOLOGY: Aplastic anemia (but most
activated T-cells) also impairs proliferation of
erythroid progenitor cells, diminishes their are in normocytic classification), chronic liver
response to erythropoietin, and decreases disease, alcoholism and other disease or
production of erythropoietin by the kidney condition that can lead to hypercholesterolemia
and reticulocytosis.
 LIVER DISEASES (chronic, obstructive jaundice),
5. HEMOGLOBIN E LCAT deficiency:
 Only hemoglobinopathies wherein its o LIVER: main organ where cholesterol is
morphology is microcytic stored and metabolized thus when
 It a structural and synthesis defect damaged cholesterol will not be
(hemoglobinopathy and thalassemia) completely metabolized leading to
 Involves same mutation as Hb C(B6) wherein hypercholesterolemia/ excess
Glu  Lysine but the point of cholesterol in plasma
mutation(substitution) occurred in 26th amino o LCAT – enzyme involve in the
acid esterification of cholesterol thus when
HEMATOLOGY 2

deficient the cholesterol(free) in the 2. MEGALOBLASTIC


plasma will accumulate leading now to  Asynchronous maturation
hypercholesterolemia  Due to abnormal development  that during
 Cholesterol in the plasma can their earliest developmental stage
be in the form of free (pronormoblast) their development is already
cholesterol or cholesterol ester abnormal  promegaloblast  2nd stage:
 The RBC membrane consists of basophilic megaloblast (b. normoblast should
phospholipid bilayer which undergo normal maturation)
have some cholesterol,  MAIN PROBLEM: Impaired DNA synthesis
proteins. CHO in the layer. The  Defective nuclear maturation caused by
cholesterol embedded in the impaired DNA synthesis (prolonged intermitotic
membrane depends on plasma resting phase; block early in mitosis)
cholesterol that when  N-C asynchrony or “maturation arrest”
cholesterol plasma is increased, o Deficient DNA synthesis (nucleus)
there will be excessive loading resulting to retarded nuclear maturation
of cholesterol into the plasma while the RNA synthesis in the cytoplasm
membrane thus enlarging the is less impeded thus cytoplasm
size of the cell. maturation proceed normally
 RETICULOCYTOSIS: any condition resulting to o While the nucleus is not yet mature, it
premature release of reticulocytes in the will not cause division but the cytoplasm
circulation keeps on maturing and enlarging
o Reticulocyte (5th in erythropoiesis) are resulting to prolonged intermitotic
immature cells seen as large cells but resting phase leading to block in early
they underwent maturation in the BM mitosis or the cell will not divide at all
o PBS: supravital stain: reticulocyte the cell become larger appearing now as
(reticulocytosis) macrocytic.
o Wright’s Stain: polychromatophilic cell  In contrast to microcytosis
(polychromatophilia) wherein the cells are small
o Shift/Stress cells: more larger because they undergo
o Released as a compensatory extradivision.
mechanism of bone marrow to the  RULE: as the cell divides, the cell
following condition: size decreases
 Acute blood loss o NUCLEUS: control center of the cell:
 Hemolysis or hemolytic anemia control all the process including the
 Bone marrow infiltration: filled division of the cell.
with abnormal cells  Not only affect RBC but all rapidly proliferating
 High level of EPO cells  enlargement of ALL rapidly proliferating
 RBC should stay in the BM for 3.5 days and 1 day cells like neutrophils
in the peripheral circulation but due to the  MCV: >100 fL; cannot be relied alone in
stated condition above, the RBC transit time in determining a megaloblastic maturation
the BM will be shorter and prematurely released  IMPORTANT FINDINGS INDICATING A
in the circulation (prolonged stay) shift/stress MEGALOBLASTIC MATURATION: MCV of >100 fL
reticulocytes + macro-ovalocyte and macropolycyte
 RBC is often seen as macro-ovalocytes (large and
oval) and Neutrophils as macropolycyte (large in
size with hypersegmentation) indication of
megaloblastic anemia.
HEMATOLOGY 2

o Neutrophil: has a normal 3-4 lobes but  Multiple Howell-Jolly bodies and many
when hypersegmented and stil has a nucleated RBCs with karyorrhexis.
normal size (9-15 um) it indicates old o DNA containing inclusion indicating
cell. nuclear remnants
o Problem in DNA synthesis which
affected the nuclear maturation thus
the nucleus is prone to fragmentation
 karyorrhexis
o Due to increase karyorrhexis
 Pancytopenia, leucopenia and
thrombocytopenia

ERYTHROKINETICS
 Total erythropoiesis is increased yet there is
reticulocytopenia indicating ineffective
erythropoiesis.
 Hyperplastic marrow: more red marrow in
order to compensate for decreased amount of
A. MACROCYTOSIS WITH NORMOBLASTIC RBCs.
MARROW o A marrow which has red
 Characterized by the presence of macrocytic hematopoietic cell (hematopoietically
cells that are not megaloblastic. This may be due active) over the marrow fats
to early release of immature erythrocytes from o Hypoplastic: more of marrow fats
the bone marrow in response to acute hemolysis o Normoplastic: normally, 50:50 ratio
of red cell and fats or 25:75 in long
and/or acute blood loss.
bones
 Not a complication but a compensation
 3x erythropoiesis
 Supravital stain: reticulocyte  Decrease M:E ratio = 1:1
 Wright’s stain: Polychromatophilic cells o Normal M:E ratio is 3:1
 Reticulocytopenia: Increased RBC in the
B. MEGALOBLASTIC ANEMIA marrow but not enough number of cells in
 Characterized by enlargement of all rapidly the circulation and due to premature lysis
proliferating cells of the body including marrow  Pancytopenia
cells. The major abnormality is the diminished
capacity for DNA synthesis, resulting to “nuclear- CHEMISTRY
cytoplasmic asynchrony” or “maturation arrest”  Related to hemolysis
RNA synthesis is less impeded than is DNA o Increase serum Lactate
synthesis, hence cytoplasmic maturation and Dehydrogenase (LD)  from RBC-
derived isoenzyme
growth continue.
o Increase bilirubin (both total and
 Most common causes: Vitamin B12 deficiency
unconjugated)
and folate deficiency o Increase endogenous carbon
NOTE: monoxide (CO)  while the
hemoglobin continuously degrade,
BLOOD PICTURE:
the heme and globin will separate
 Presence of macroovalocytes and giant
and heme will further separate into
hypersegmented neutrophils.
iron and protoporphyrin IX.
 Basophilic stippling: precipitated ribosomes
Protoporphyrin IX will further
indicating a disturbed erythropoiesis and
undergo degradation releasing
erythrocyte metabolism
biliverdin and CO. In normal
HEMATOLOGY 2

hemolysis, carbon monoxide is


exhaled but if hemolysis is increased
CO is also increased.
o Common source of CO is exogenous:
factory and automobile exhaust
 Others: different mechanism
o Increased serum muramidase 
present in granules of neutrophils
 Disturbed granulopoiesis
thus enlarged and
hypersegmented neutrophils
and easily damaged thus
releasing their enzyme
content (muramidase).
o Increased homocysteine  due to
failure convert into methionine
 Homocysteine should receive
methyl from folate to
convert into methionine, but C. CYANOCOBALAMIN (VITAMIN B12) DEFICIENCY
due to folate deficiency it  Not common to occur because the body’s daily
fails to be converted. requirement of B12 is very minimal (around 2-5
DNA = double stranded helix consisting of sugar- ug/day)
phosphate backbone and nucleotides.  When intake is stopped, this type of anemia can
be acquired after 3-6 months or a year. Because
In the formation of thymidine structure to thymine, the body’s utilization of B12 is also minimal thus
both folate and B12 are needed. Therefore deficiency B12 deficiency takes gradually.
in folate and B12 will result to impaired DNA synthesis.
 Vitamin B12 is required in the Demethylation of
The bone marrow converts
folate during DNA synthesis
deoxyuridinemonophosphate into
deoxythymidinemonophosphate using the enzyme  Folate is absorbed through the ileum with the aid
thymidylate synthethase which needs a cofactor of the intrinsic factor (IF) or Castle’s factor
tetrahydrofolate (THF) which is from dietary folate. produced by the parietal cells, and is transported
Dietary folate enters the plasma in the form of 5- by transcobalamin II (TC II) in the plasma.
methyl THF and enters again the red cell in the form of o Intrinsic factor: a protein synthesized by
5- methyl THF but before it could participate in the gastric mucosa; without it, B12 can’t be
synthesis of DNA structure, it needs to undergo absorbed
demethylation wherein there should be removal of o In ileum, cells (enterocyte) with
the methyl group. The methyl removed will be receptors for B12 are available however
transferred to homocysteine thus converting it into it is specific only to B12-IF complex. Thus
methionine by the enzyme methionine synthase
when only B12 is pass through it, it will
needing the cofactor B12.
not be recognized therefore not
TWO MOST COMMON CAUSES OF MEGALOBLASTIC absorbed.
ANEMIA: o Transcoblamin II: transports B12 to sites
1. B12 deficiency anemia where it could be metabolized or stored
2. Folate deficiency anemia like in the liver, bone marrow and other
tissues.
 RDA: 2-5 ug/day
 TWO SOURCES OF B12:
HEMATOLOGY 2

o DIET: primarily from food of animal  Inadequate myelin synthesis: neurologic


origin  main source of B12 damage
o LARGE INTESTINE: synthesized by some  Hyperhomocysteinemia: cardiovascular
of the normal flora in the intestine disease.
(lactobacilli spp. and bacteroides spp.). NOTE:
However the absorption of B12 is in the RELATED CAUSES IF VITAMIN B12 DEFICIENCY:
ileum but the production comes from 1. INADEQUATE INTAKE
the large intestine. Thus, B12  RDA: 2-5 ug/day from meat diet
synthesized in the large intestine is not  Common in strict vegan people
all/readily available for consumption of
2. MALABSORPTION SYNDROME
the human body.
 Any condition affecting the ileum
NOTE: (inflammation): Celiac disease, ileal disease
ACQUISITION OF METABOLISM OF B12 (Crohn’s disease/regional enteritis) tropical
 B12 comes from the diet (meat origin) once sprue, resection of the small bowel
reaches the stomach, the gastric juice is very  Imerslund-Grasbeck syndrome – deficiency
acidic because of the HCl produced by the or absence of IF-B12 receptors
parietal cells. Thus B12 can be damaged. In  Zollinger-Ellison syndrome – hypersecretion
order to protect B12 from acid destruction, syndrome  gastric hypersecretion (acid and
salivary glands produce haptocorrin or juices) that can degrade B12 and prevent it
Transcobalamin I (TC I). While masticating the from binding with IF and therefore B12 is not
food, B12 combines with TC I. Since TC I absorbed
protects B12, it can bypass the acidity and
digestive action of the stomach in small 3. LACK OF AVAILABILITY OF COBALAMIN
intestine (duodenum;1st portion) the pH of  Blind loop syndrome
the chyme is neutral to alkaline thereby o Refers to bacterial overgrowth
destroying haptocorrin and B12 will be o The large intestine (colon) is rich in
released. B12 combines with Intrinsic factor normal flora that synthesis vitamins
(IF) coming from the parietal cells of the but in this case, what will proliferate
stomach and thus forming the B12-IF in the LI are the abnormal flora which
complex. The complex goes down until the will utilize the available B12 at the
jejunum and terminal part – ileum where it expense of the host. And this
will be absorbed by the intestine. Once abnormal flora will also destroy the
absorbed, IF will be detached and B12 will be normal flora in the expense of their
released into the circulation. B12 will then capacity of producing some
combine with Transcobalamin II and be important vitamins (B12)
brought into the liver (for storage or
metabolism), bone marrow (for utilization and  Infection with Diphyllobothrium latum
DNA synthesis) and other tissues (for o D. latum is the longest tapeworm
utilization being a cofactor). infecting human which resides in the
ileum
IMPORTANCE OF B12 o Bothriocephalus anemia: long
 For metabolism tapeworm that competes with the
 Formation of RBCs host with the absorption of B12 and
 Maintenance of CNS (brain and spinal cord) secretes substance that inhibit
binding of IF with B12.
CONSEQUENCES OF B12 DEFICIENCY
 Ineffective DNA synthesis: Megaloblastic 4. PRESENCE OF AUTOANTIBODIES
anemia  Can be either those who destroy the intrinsic
factor or those which destroys cells(parietal)
HEMATOLOGY 2

that produce IF  Inability of the gastric cells become defective and can’t
mucosa to secrete IF (Pernicious anemia) produce HCl
 Achloridria: absence of HCl;
5. DRUGS/ SUBSTABCE-RELATED more specific of pernicious
 e.g. Neomycin and ethanol  chronic anemia especially when the
alcoholics autoantibodies destroy the
parietal cells.
6. ABNORMAL PRODUCTION OF TC-II
 HCl: important in digestion in
the stomach  digestion of
D. PERNICIOUS ANEMIA proteins
 Autoimmune type of vitamin B12 deficiency o Episodic abdominal pain, constipation,
anemia and diarrhea
 Types of Autoantibodies: o Symptoms of anemia with a
o Anti-parietal cell antibodies: destroys combination of skin pallor and lemon-
parietal cells yellow skin
 Intrinsic factor: complexes with o Diffuse and irregular degeneration of
B12 in order for it to be the white matter of the CNS
absorbed  Memory loss, numbness and
 HCl: gives acidity of the tingling in toes and fingers
stomach  absence can lead to damaged nerves
achloridria  Symmetric sensation of “pins &
needles” of the distal
o Anti-intrinsic factor antibodies extremities
 IF production is normal, but IF is  In advance cases, brain may be
inhibited by the autoantibodies affected resulting to irritability,
thus it cannot bind with B12  emotional instability, or a
non-absorption of B12 change in personality
 2 TYPES OF ANTI-INTRINSIC (megaloblastic madness)
FACTOR ANTIBODY:
NOTE: DIAGNOSIS OF VITAMIN B12 DEFICIENCY
Blocking antibody: blocks the
 Normal plasma vit. B12 levels: 180-914 ng/L
binding of cobalamin to IF
Binding antibody: binds to the 1. SERUM COBALAMIN ASSAY
cobalamin-IF complex  A microbiological assay using an organism (E.
becoming trimolecular complex gracilis) that requires B12 for its growth.
receptors will not recognize;  Organisms used: Euglena gracilis and
and prevents the complex from Lactobacillus leichmannii
binding to receptors in the  Patient’s plasma + organism incubated for
ileum 48-72 hours at 37 degree Celsius
 Clinical signs & symptoms include:  If the patient’s plasma has a normal B12, it will
o Atrophic glossitis: tongue will atrophy support the growth of the organisms
and become inflamed due easy organisms will grow and cause turbidity of the
desquamation of surface epithelial cells medium
o If the patient’s plasma has an
(lack of integrity because of B12 def.) 
abnormal B12  growth will be
beefy red tongue
retarded  organisms will not grow
o Atrophic gastritis and achloridria:
and will not cause turbidity
leading to malabsorption; when o TURBIDITY WILL BE COMPARED WITH
stomach become inflamed, parietal STANDARD CONTROLS
HEMATOLOGY 2

 Highly sensitive but it is lengthy to performed binding with the reagent B-


but; lactoglobulin
o Not sensitive to patients taking o Differentiation: using the radioassay.
antibiotics  antibiotics are in plasma
thus when incubated with the 5. SCHILLING’S TEST
organisms, it can destroy or kill these  Specific test for Pernicious Anemia
thus this test should not be done with  Provides a measure of the body’s ability to
patients undergoing an antibiotic secrete viable IF & absorb orally administered
therapy Co-labeled B12.
 Reference value: 200-900 mg/dL  PART ONE: patient is given a physiologic dose
of B12 labeled with radioactive cobalt (0.5 -2
2. METHYLMALONIC ACID & HOMOCYSTEINE ug) ingested orally to see whether the patient
ASSAY can absorb the B12 and after 1-2 hours;
 B12 is a cofactor of methymalony CoA mutase another flushing dose is given  injection of
in the isomerization of methylmalonyl CoA to non-radioactive vitamin B12 (1000 ug)
Succinyl CoA. Deficiency of B12 will lead to intramuscularly in order to saturate the
excretion of increased amount of binding sites of B12 so that the minimal
methylmalonate in the urine. amount of B12 absorbed will not bind in the
 Methyl malonyl CoA  MMCoA mutase with binding site instead it will be urinated. check
cofactor Vit. B12  Succinyl CoA B12 levels in the patient’s urine
 Measured by: o Patients with IF or NO pernicious
o Methyl malonic acid assay: GC-MS anemia: the cobalt labeled B12 given
o Homocysteine assay: GC-MS; HPLC; orally in the duodenum will combine
Fluorescence Polarization with IF available in the patient to now
 In both B12 and folate deficiency, allow B12 absorption. B12 absorbed
homocysteine will increase.  homocysteine will proceed to the plasma and those
assay some will be stored in the liver,
plasma protein binding and urinary
3. DEOXYURIDINE SUPPRESSION TEST excretion.
 Indirect test that measures the ability of the  Patients with normal IF: excrete >7% ingested
bone marrow cells in vitro to utilize B12
deoxyuridine in DNA synthesis.  If the first part of the test is normal: excreted
 It is abnormal in both cobalamin (B12) and >7% B12  report it as NORMAL. But if there
folate deficiency. is abnormal result (<7% or none excreted) that
may be due to deficient or absent IF 
4. COMPETITIVE PROTEIN BINDING proceed to PART II
RADIOASSAY o If IF is absent, there will be no
 Method of choice for both B12 and folate intestinal absorption of B12 thus,
deficiency quantitation ingested B12 will be excreted through
 Principle: patient’s B12 and folate compete defecation.
with Co-B12 and -folate in binding with hog IF  PART TWO: Done after 24 hours of the
and B-lactoglobulin conduct of first part of the test. Patient is
 Patient’s B12  hog IF  Co-B12 given an oral cobalt-labeled B12 with IF that
o Competition between the patient’s will allow absorption of B12 in the ileum.
B12 against the cobalt-labeled B12 in Result is therefore expected to be impoved.
binding with hog IF o Urinary excretion: improved >7% or
o Differentiation: using the radioassay 15%  PERNICIOUS ANEMIA
 Patient’s folate  B-lactoglobulin  I-folate o Low cobalt in urine  Other B12
o Competition between patient’s folate deficiency anemia
against the iodine-labeled folate in
HEMATOLOGY 2

OTHER TESTS RELATED CAUSES IF FOLATE DEFICIENCY


1. TEST FOR AUTOANTIBODIES  Inadequate intake/Dietary deficiency
 For pernicious anemia  Liver disease
 Identify serologically what specific antibody is  Alcoholism: alcoholic is hepatotoxic
produced  Increased demand/requirement
 More specific than schilling’s o Prone to babies and pregnant women
 Defective absorption
2. GASTRIC ANALYSIS (HCl) AND SERUM  Inadequate utilization of folate
GASTRIN  Presence of folate antagonists (antimalarial
 For pernicious anemia drugs that inhibits utilization of folate)
 HCl: Achloridria due to damage in parietal
cells DIAGNOSIS OF FOLATE DEFICIANCY
 Gastrin: peptide hormone secreted by the 1. Microbiologic assay using Lactobacillus casei
gastric walls which functions to stimulate  Serum and Red Cell folate: usually < 3 ug/L
stomach to secrete juice and acids. Since there
is decrease HCl, gastrin will be stimulated to 2. Deoxyuridine Suppression Test
induce more HCl production. Although serum  Ability of BM to utilize deoxyuridine into
gastrin is plenty, there will be still no HCl since deoxythymidine and finally into DNA
the parietal cells are destroyed. structure
 DECREASE HCl and INCREASE SERUM GASTRIN
3. Quantitation of Vitamin B12 and folate
3. SERUM HOLOTRANSCOBALAMIN ASSAY through competitive protein binding
 Complementary test radioassay
 Holotranscobalamin: metabolically active  Method of choice
form of cobalamin (B12)
4. Homocysteine Assay
4. FECALYSIS (FOR BOTHRICEOHALUY ANEMIA)
 Operculated eggs of D. latum
F. TROPICAL SPRUE & GLUTEN-SENSITIVE
ENTEROPATHY
 A condition that can cause both vitamin B12 and
E. FOLIC ACID/ PTEROYLGLUTAMIC ACID folic acid malabsorption as a result of intestinal
DEFICIENCY atrophy.
 Features are similar to Vitamin B12 deficiency  Intestinal atrophy (SI and proximal intestine)
but no neurologic symptoms; leukopenia and Vit. B12 and folic acid malabsorption
thrombocytopenia are less constant o Tropical Sprue: intestinal atrophy of the
 RDA: 50 ug folic acid or 400 ug food folate small intestine
 The clinical s/s of it is very similar with B12 o Gluten-sensitive enteropathy: intestinal
deficiency except NO NEUROLOGIC SYMPTOMS atrophy of the proximal intestine
 uncommon to occur even in severe cases
 Absorption: small intestine: duodenum and NOTE: Tropical Sprue & Gluten-sensitive enteropathy
jejunum (same with iron absorption) THERAPY: depends on the condition
1. ORAL COBALAMIN
o Small intestine parts: 1st duodenum 
 B12 deficiency: B12 supplement
2nd jejunum  3rd ileum (B12
absroption)
2. PARENTERAL CYANOCOBALAMIN
 Deficiency inhibits the thymidine  For pernicious anemia  through IV/
 SOURCE: vegetables and poultry products (eggs) injection
NOTE:
3. ORAL FOLATE
HEMATOLOGY 2

 Folate deficiency: Folate supplement o HYPOPLASTIC: preferred term meaning


severe depletion of blood cells
o APLASTIC: may be confused due to the
NOTE: RECAP prefix “a” meaning absence; but in
DEVELOPMENTAL DEFECT:
anemia it means the same as hypoplastic
MICROCYTIC: abnormal cytoplasm maturation (hb
 Clinical signs include the common signs of
synthesis)
MACROCYTIC: Megaloblastic: Asynchronous anemia plus infection and bleeding seen also in
maturation due to defective DNA synthesis leukemia
 Splenomegaly & lymphadenopathy are ABSENT
in aplastic anemia but present in leukemia
NORMOCYTIC-NORMOCHROMIC ANEMIAS o In leukemia, there are many leukemic
 Synchronous maturation: not a developmental cells that causes the spleen to be
defect but a survival or proliferation problem overworked (remove cells) 
 Anemia occurs due to: splenomegaly.
o Increased destruction  Hemolytic o For ALL, the lymph nodes are also
anemia: can be INTRINSIC or EXTRINSIC infiltrated  lymphadenopathy
 Problem in the
circulation/spleen wherein a
lysis occur
 Increased in reticulocyte count
due to compensatory
mechanism of a normal bone
marrow
NORMAL BM HYPOCELLULAR HYPERCELLUL
o Decreased in proliferation or
BM AR BM
hypoproliferation 50% of red Decrease in red Increase in red
 Bone marrow defect wherein marrow and marrow but marrow but
there is decrease production of 50% of marrow increase in white decrease in
cells fats marrow(fats) white marrow
 Decreased or normal (fats)
reticulocyte count due to bone Ratio depends Seen in
marrow failure on age: hypoplastic/aplas Seen in
 Differentiated through reticulocyte count Younger child: tic anemia polycythemia
100% red vera
I. ANEMIA OF BONE MARROW FAILURE marrow
Adults: 50:50;
 Could be due defect of bone marrow cells
25:75
(damaged)
 Abnormal or lack of stimulation of the hormone
(EPO) even with normal bone marrow PATHOGENESIS
1. Direct damage to the hematopoietic
A. APLASTIC/ HYPOPLASTIC ANEMIA stem & progenitor cells decreased in
 Characterized by a severe reduction of the blood cell count
amount of hematopoietic tissues that results to o Causes: ionizing radiation, toxic
deficient production of blood cells (hypocellular chemicals (benzene)
bone marrow) o Infiltration BM: myelophthistic
anemia
 BONE MARROW: hypocellular
o competition for nutrients by
 CIRCULATION: pancytopenia
normal and abnormal cells
cancerous cells take up the
HEMATOLOGY 2

oxygen and nutrient supply  Associated with other group of congenital


o PANCYTOPENIA anomalies or with genetic predisposition to
2. Immune-mediated chronic bone marrow failure. Predisposition to
cancer and chronic bone marrow failure.
destruction of marrow cells  Patient is born without aplastic anemia but has
o Antibody destruction wherein
congenital genetic abnormality that made him
patient produce autoantibody
predispose to suffering other
that damages its own cell.
o PANCYTOPENIA malignancies/cancer.

3. Low effectiveness of EPO


FANCONI’S ANEMIA 9CONGENITAL APLASTIC ANEMIA)
(cannot activate erythroid
stem cell)  Congenital bone marrow defect that leads to
o Defective/inefficient hormone decrease production blood cells
that causes insufficient  Hereditary Disorder: Autosomal recessive trait
stimulation of the bone marrow or pattern of inheritance
o PURE RED CELL APLASIA  Chromosomal defect are found in lymphocytes
4. Low production of EPO like and BM cells.
 With features of developmental abnormalities
in renal diseases that include hyperpigmentation, short stature
o Normal function of EPO but low
(inability to grow), hypogonadism, mental
in number thus unable to
retardation, strabismus (cross-eyed) and
stimulate the bone marrow
malformation of the extremities.
o PURE RED CELL APLASIA
 Fanconi’s syndrome: different and has no
relationship from this kind of anemia
o Defect in PCT reabsorption thus it is a
A.1. IDIOPATHIC APLASTIC ANEMIA renal tubular defect wherein the
important solute in the filtrate are not
 The cause is of unknown etiology
reabsorbed.
 The development of sign and symptoms is
hideous and gradually occurring until it becomes FAMILIAL APLASTIC ANEMIA
a full-blown anemia
 Subset of Fanconi’s anemia.
A.2. ACQUIRED APLASTIC ANEMIA  SCHWACHMAN – Diamond syndrome prone to
develop familial aplastic anemia
 May be the effects of exposure to certain
physical and chemical agents some drugs like  Patients may have pancytopenia and a
antimicrobials, anticonvulsants analgesics hypocellular marrow without major
developmental abnormalities.
 SECONDARY TO:
o Ionizing radiation  high doses
damages bone marrow A.4. PURE RED CELL PLASMA
o Benzene  chemical that has toxic
effect to BM  Benzene Hypersensitive  Only the red cell are decreased
Aplastic Anemia NOTE:
o Chloramphenicol  most common
antibiotic causing AML and Aplasic 1. Transitory Arrest of Erythropoiesis/
anemia  toxic to BM transient aplastic crises – may occur during
the course of a hemolytic anemia often
preceded by an infection with parvovirus B19
A.3. CONSTITUTIONAL APLASTIC ANEMIA
HEMATOLOGY 2

infects CFU-E  In chronic disorders/inflammation: WBC are


2. Transient Erythroblastopenia of Childhood – activated causing them to release cytokines.
associated with a history of viral infection Increased cytokines (interleukins, TNF,
within the last 3 months (humoral inhibition) interferons)  alter iron metabolism that affects
3. Acquired Pure Red Cell Aplasia – associated hb synthesis (microcytic – hypochromic) but if
with thymoma the effect of it is on blood cell production like will
4. Congenital Red Cell Aplasia/ cause decreased EPO because it inhibits kidneys
or it causes decrease in RBC production as it
Congenital Hypoplastic Anemia directly suppresses the erythroid progenitors
(Diamond-Blackfan anemia) (normocytic - normochromic)
 Defects in BFU-E and CFU-E with  Increased cytokines and Decreased EPO
accelerated apoptosis (programmed cell
ANEMIA OF RENAL INSUFFICIENCY
death) thus decrease production of RBCs
o Progenitor cells BFU-E CFU-E  Normocytic anemia due to renal disease wherein
that give rise to pronormoblast the kidney will not be able to produce enough
mature erythrocytes hormones (EPO) which is the main growth factor
 Most common type caused by a defect in for erythropoiesis
the erythroid-committed progenitor cell
 Decreased EPO (inefficient stimulation)
decreased BM response thus decreased RBCs
II. MYELOPLASTIC ANEMIA  Patients on dialysis is often given commercially
 Associated with marrow replacement by produced EPO and undergo frequent transfusion
metastatic carcinoma, multiple myeloma, of packed red blood cells
leukemia or other conditions.
ANEMIA OF ENDOCRINE DISEASE
 This is characterized by the presence of varying
number of normoblasts and immature  EPO has hormonal regulation and its function in
neutrophils. (leukoerythroblastic-anemia) stimulating blood production is also affected by
o Leukoerythroblastic reaction: increase in hormones produced by the endocrine glands like
immature granulocytes or myeloblast the following:
and immature erythrocyte. o Thyroid, pituitary, adrenal, gonads
o Occasionally macrocytic but more
ANEMIA IN LIVER DISEASE
commonly normocytic-normochromic
 Marrow replacement: space occupied by cancer  More characterized by macrocytic anemia but
cells  overcrowding and consumption of occasionally may be normocytic
nutrients and oxygen supply at the expense of  Liver diseases are associated with abnormal
normal cells by the cancer cells metabolism and storage of folate
 In leukemia, there is decrease RBC (anemia), megaloblastic anemia
decrease WBC (infection) and decrease platelets
(bleeding tendencies). Anemia that develops in
leukemia is Normocytic-Normochromic. IV. CONGENICAL DYSERYTHROPOIETIC
ANEMIA (CDA)
III. ANEMIAS OF SYSTEMIC DISORDERS  Group of anemia: inherited refractory anemia
(ASSOCIATED WITH CHRONIC DISEASES) characterized by erythroid multinuclearity,
 These include anemia of renal insufficiency, karyorrhexis and bizarre malformations.
anemia in liver diseases, and anemia of o Refractory means unresponsiveness to
endocrine function, among others. stimulation and medication
 RBCs are usually normocytic-normochromic,
occasionally microcytic-hypochromic.
HEMATOLOGY 2

NOTE: o Intrinsic defects (defects of the red cell


itself)
o Abnormal membrane of RBCs:
Hereditary spherocytosis, elliptocytosis,
South-east asian ovalocytosis
o Abnormal metabolism/ metabolic
disorder: embden-meyerhof pathway or
hexose-monophosphate shunt is not
occurring normally due to enzyme
deficiency.
TYPE I: Megaloblastic changes with some binuclearity o Hemoglobin defects: RBCs are
 has nuclear bridging. synthesizing abnormal hemoglobin that
 More on macrocytic. makes the cell prone to lysis.
 Defective gene (Ch 15) o Most are inherited defects
TYPE II: HEMPAS (Hereditary Erythroblastic  Extrinsic defects (factors outside the red cell)
Multinuclearity with a Positive Acid Serum Test) o Normal RBC but the environment of it is
abnormal.
TYPE III: Giant erythroid precursor with more o Damaged blood vessels/
pronounced multinuclearity,
microcirculation
 More on macrocytic
o Plasma has antibodies or toxic agents
 Defective gene (Ch 15)
that can destroy the cells
o Most are acquired defects
V. APLASTIC ANEMIA ASSOCIATED WITH
CHEMICAL OR PHYSICAL AGENTS
 Chemical Agents: Benzene  toxic to BM
causing Toxic Aplastic Anemia and
Chloramphenicol
 Physical Agent: Ionizing Radiation  RBC (most
sensitive)  WBC  Platelets (more resistant;
last to be affective)

HEMOLYTIC ANEMIAS
 Belong to normochromic-normocytic anemia
 Normally, 1% of our RBCs are destroyed daily by
extravascular hemolysis
 Hemolysis can occur:
o Extravascular hemolysis: outside the
blood vessels  spleen (splenic
macrophages)
o Intravascular hemolysis: within
the blood vessel or circulation
o In both cases, there will be the release of
hemoglobin that may cause the loss of it.
Hemoglobin contains most of the body’s
iron (60% of total iron)
 Increased red cell destruction can be cause by:
HEMATOLOGY 2

If the iron released from the EXTRAVASCULAR HEMOLYSIS occuring in macrophages. When the RBCs are
hemoglobin is not recycled, it will abnormal, the spleen function in filtering the blood and removes any abnormal or
be loss. The body’s mechanism in senescent/old red cells called splenic culling.When there are alot of abnormal cells,
order to recycle the iron: Our splenic culling/ extravascular hemolysis will be increased resulting to the release of
iron source may come from the hemoglobin due to hemolysis of RBCs. Hemoglobin undergoes degradation by
diet and recycled iron, thus it is separating into heme and globin. Globin will be recycled as amino acid and goes
important for our body is able to back to cytoplasm and plasma as part of of the amino acid pool for further protein
recycle iron. When RBCs are synthesis. Heme undergoes further degradation releasing iron and recycled.
hemolyzed, it will release Protoporphyrin IX will also be released from the heme and be broken down into
hemoglobin and it will be endogenous carbon monoxide (exhaled) and biliverdin. Biliverdin is reduced into
degraded to release the iron. bilirubin (unconjugated) and released into the plasma. So what increases in the
Iron can go directly to cells for plasma is the unconjugated type which is water-insoluble and is immediately bound
reuse, or can be bound to to albumin. Unconjugated bilirubin can’t be excreted in the urine because kidney is
transferrin for transport or not able to filter it due to the following reasons: it is water-insoluble and they are
stored in the form of ferritin and bound to albumin (HMW). This albumin-unconjugated bilirubin complex is brought
hemosiderin. into the liver for conjugation. It is conjugated with 2 molecules of glucuronic acid
catalyzed by the enzyme UDP-glucuronyl transferase to form the conjugated
bilirubin called bilirubin diglucuronide. Conjugated bilirubin is stored in the gall
bladder and when needed it forms part of the bile. When needed for digestion it
will be secreted along with the bile into the small intestine to be acted on by some
bacterial enzyme and intestinal enzymes thus it converts into urobilinogen. ½ of the
urobilinogen will reabsorbed going back to the circulation then into the liver. The
other half will be converted into urobilin and stercobilin to be excreted in the stool.
FINDINGS: Increased plasma bilirubin, increased urine urobilinogen, increased
endogenous carbon monoxide.

INTRAVASCULAR HEMOLYSIS: The red cell is not engulf but it lyses in the circulation
releasing now hemoglobin in the circulation resulting to free hemoglobin in the
plasma. Hemoglobin is a LMW protein thus it can be easily filtered (solutes with MW
of <70,000 daltons) by the kidney and excreted into the urine resulting to
unpreservation of the hemoglobin content like iron. To prevent haemoglobin loss,
haptoglobin from the liver will bind with it forming now the hemoglobin-
haptoglobin complex (HMW) and direct into liver for degradation. But due to this,
haptoglobin is consumed. When there is continuous hemolysis, haptoglobin will be
depleted. Thus, hemoglobin becomes free again and will be degraded into heme
and globin in the plasma. Some heme will be oxidized and binds with albumin
forming methemalbumin and some will also bind with hemopexin forming heme-
hemopexin complex and these two will be brought back in to the liver for
degradation.
FINDINGS: hemoglobinuria or methemoglobinuria; Decreased haptoglobin;
increased plasma methemalbumin; increased carbon monoxide
HEMATOLOGY 2

TYPES OF HEMOLYSIS (intravascular hemolysis)


 Increased urine urobilinogen
 EXTRAVASCULAR (MACROPHAGE-MEDIATED):
(extravascular hemolysis)
destruction of senescent red cells by splenic
macrophages. RBC Survival Studies (Radioactive Chromium
 INTRAVASCULAR DESTRUCTION (MECHANICAL (51Cr)
HEMOLYSIS): may be caused by the turbulent o RBC half-life: 25-32 days
environment in the circulation o Cr-RBCs IV blood count every 1-2 days
for 10-14 days
3 MECHANISMS ON HOW THE BODY CONSERVES IRON o Residual activity: index of the
IN HGB DURING INTRAVASCULAR HEMOLYSIS intravascular lifespan
1. Haptoglobin binds with free Hb preventing it o Detects sites of red cell destruction
from being filtered by the kidneys
2. When haptoglobin is depleted, heme is liberated
from hb and binds with hemopexin.
3. Oxidized heme binds with albumin forming
Methemalbumin

LABORATORY FINDINGS: RBCs and HgB Destruction


HEMATOLOGY:
 Reticulocyte count measured by RPI
o Reticulocytosis/
polychromatophilia
 Decreased red cell hemoglobin, Hct, and
RBC count
 Normocytic and/or occasionally
Macrocytic anemia representing the
reticulocytosis that is prematurely
released in the circulation

BONE MARROW
 Erythroid hyperplasia (decreased M:E)
 Increased Plasma hemoglobin and
bilirubin (unconjugated)
o Intravascular: highly increased
o Extravascular: slightly increased
 Decreased Serum haptoglobin
o Intravascular: severely decreased
to 0
o Extravascular: slightly decreased
 Increased plasma methemalbumin 
formed when haptoglobin can’t no longer
support hemoglobin
 Increased LHD due to hemolysis 
increased in isoenzyme LD 1 and 2

URINALYSIS
 Hemoglobinuria and methemoglobinuria
HEMATOLOGY 2

TEST PARAMETER INTRAVASCULAR HEMOLYSIS EXTRAVASCULAR


HEMOLYSIS
H Hb, Hct, RBC count Decreased Decreased
E
M Reticulocyte count Increased Increased
A
T Bone Erythroid hyperplasia Erythroid
O marrow (decreased M:E ratio) hyperplas
L examinatio ia
O n (decrease
G d M:E
Y ratio)
RBC morphology Normocytic some macrocytic Normocytic some
macrocytic

P Free hemoglobin Increased Slightly increased


L
A Bilirubin Increased Unconjugated Increased
S Unconjugated
M
Haptoglobin Decreased/Absent Slightly decreased
A
C
H Lactate dehydrogenase Increased Slightly increased
E
M
Hemopexin Decreased Slightly decreased
I
S Endogenous CO Increased Increased
T
R
Y

U Free hemoglobin Positive Negative


R
I Urobilinogen Increased Increased
N
E Hemosiderin Positive Negative

1. HEREDITARY SPHEROCYTOSIS  It may involve a defect or deficiency in spectrin


 A defect in the RBC membrane protein or a defect in the binding of spectrin to protein
composition leading to hyperpermeability of the 4.1, ankyrin, band 3, beta-spectrin, protein 4.2.
cell to sodium. o Proteins involves are vertical proteins
 A genetic problem that leads to production of that support the red cell membrane.
abnormal membrane protein or cause non-/ or o Many proteins can be involve:
deficient production of red cell protein. combination of which or it could also
 Repeated removal of parts of the unsupported mean the interaction of these proteins
membrane results to spherocyte formation. to together like the connection of
 Loss of unsupported lipid membrane spectrin to protein bands.
HEMATOLOGY 2

 Uncoupling of unsupported LABORATORY FINDINGS:


phospholipid bilayer due FINDINGS IN BOTH HEREDITARY AND ACQUIRED
defective protein. The detached  Spherocytes, microspherocytes and many
lipids will form vesicles and reticulocytes
remaining part becomes smaller o Seen in both hereditary and acquired
cell without a central pallor condition
spherocytes. (HEREDITARY) o Reticulocytes  as compensatory
mechanism of BM
 Spherocyte does not always mean a hereditary
 Osmotic Fragility Test – always increased
condition, it could also be an acquired condition
o Since the spherocytes are non-
(AUTOIMMUNE ANEMIA that can lead to deformable because they are already
spherocytosis). fully distended, they are fragile and
 It could also be ARTEFACTUAL: in an old stored easily lyse.
blood, most of the red blood cells are  MCV: normal/slightly decrease
spherocytes. o Normocyte is larger than spherocyte,
 Hence, it is needed to differentiate the true but when it comes to volume, they are
hereditary membrane defect (HS) from other the same. The volume of spherocyte is
causes of spherocytosis like in immune closely similar to normocyte despite
hemolytic anemia. the difference in size because
o IMMUNE HEMOLYTIC ANEMIA: although the spherocyte is smaller, its
entire volume is occupied by the
antibody sensitized the red blood cell. So
hemoglobin (no central pallor) fully
when this sensitized RBCs passes
distended. Unlike the normocyte
through the spleen, the macrophages
wherein it may have a normal size but
will recognize it as a foreign/abnormal its volume is not entirely occupied
cell and remove the inclusion (attached (1/3 central pallor).  SPHEROCYTE
antibody) in the cell (splenic pitting). As and NORMOCYTE have the SAME
the macrophage pulls off the antibodies VOLUME.
from the red cell, it also knits-off some o DIFFERENCE: in SIZE  spherocyte
parts of the red cell. The remaining RBC (smaller) vs. normocyte (larger)
fragments will reseal and form into
spherocytes.  MCHC is increased  no central pallor due to
 Since some of these proteins take part in ion- diminished volume thus the entire volume will
exchange channel, deficiency will cause be occupied by the hemoglobin. When
hemoglobin is measured in relation to the cell
alteration or have defect on the channel thus
volume, it is high. Hence, MCHC is above 36%.
ion transport is affected.
o Spherocytes are cells with highest
o Sodium-Potassium ATPase: instead of MCHC but the volume is normal
normal ion-exchange (normal amount  Autohemolysis test – positive
of Na (major extracellular cation) and K o When the spherocytes are incubated
(major intracellular cation) is for 48 hours, the more they become
maintained.  Defect in transport  sphere therefore they hemolyse on
will result to accumulation of sodium their own.
inside or the cell gains more sodium
causing also the cell to gain more water DIFFERENTIATION:
hence the cell will be bloated.  Direct Antiglobulin/Coomb’s Test (DAT/DCT)
– negative
o Test done in blood banking to
determine occurrence of in vivo
sensitization of RBCs
HEMATOLOGY 2

o Determine whether the RBCs were whether the spherocytosis is hereditary,


attached with antibodies in vivo immune or other causes.
o In HEREDITARY: no antibody-  For hereditary spherocytosis and other
associated whether in vivo or in vitro anemia characterized by spherocytosis
 NEGATIVE in DAT o Spherocytes are test using OFT
o In IMMUNE HEMOLYSIS that result to because they are the most fragile cell
SPHEROCYTOSIS: there are  able to detect the presence of
antibodies-associated  POSITIVE in spherocyte in the circulation.
DAT o RESULT IN HS: 0.60 - 0.65% saline
CLINICAL SIGNS AND SYMPTOMS solution, hemolysis is already
JAUNDICE AND SPLENOMEGALY observed: spherocyte (fragile) easily
 Since it is a type of hemolytic anemia, increase lyse
in bilirubin causing the jaundice.  PRINCIPLE: The cell is placed in a hypotonic
 With the predominance of spherocytes in the saline to cause osmosis (the shift of the fluid
circulation, the spleen becomes overworked from the hypotonic saline into the red blood
in the response of removing them.  cell). If the red cell could still contain its
SPLENOMEGALY: enlargement of the spleen volume, the RBC would be able to absorb
 When the spleen is enlarged, the more it water without lysing and if not, it will easily
sequesters blood and destroys more blood. hemolyse. POSITIVE RESULT: Hemolysis
 HEREDITARY SPHEROCYTOSIS (HS): the main  RESULTS:
cause of hemolysis is the spleen. It is the o Incomplete hemolysis: 0.45%
spleen that causes the hemolytic process  Faint pink tinge supernatant
because the function of the spleen is to filter with intact red cell button
the blood thus it removes abnormal cells. o Complete hemolysis: 0.35%
Since spherocytes are abnormal cells, they  Homogenous pink/red
remove it through hemolysis. solution with the absence of
 It is the hemolytic process that will lead to the red cell button.
complications  when the bilirubin levels  Fragility means increased OFT  cells are
increased and the hemoglobin is rearbsorbed easily lyse (spherocytes)
in the kidneys wherein the heme is toxic to the  INCREASED OFT:
kidney  HS. o HS, abnormal membrane, severe G-6-
TREATMENT PDH deficiency, PK deficiency,
SPLENECTOMY is considered to Hemolytic anemia
prevent/lessen/correct the extravascular hemolysis  DECREASED OFT:
however it will not correct the spherocytosis because o Resistant to red cell lysis even when
it is hereditary and therefore the abnormal gene will placed in hypotonic solution
always be there causing the production of  With increased surface area-
spherocytes. But the spherocytes will no longer to volume ratio
hemolyse because the spleen was removed.  Hypochromic cells, target
cells/leptocytes, sickle cell
o Severe IDA (microcytic-hypochromic
LABORATORY TESTS: cell)
1. OSMOTIC FRAGILITY TEST o Thalassemia (microcytic-
 A test to identify the ability of red cells to hypochromic and target cell)
absorb water without lysing. o Sickle cell anemia: If the sickling is still
 It reflects the shape and size of erythrocyte mild and is reversible, when the sickle
(specifically the surface area-to-volume ratio) cell absorbs water it goes back to
 Not a confirmatory test for HS; it will only being a normal cell. However, if its
identify the presence of fragile cells like membrane is totally damaged, the
spherocytes. But it will not determine
HEMATOLOGY 2

sickling becomes irreversible the tube corresponds to the


therefore becomes resistant. drops of 0.5% NaCl)
 AFFECTED BY: 3. Using the same dropper,
o Surface area-to-volume ratio add distilled water into each
 A normocyte with normal tube until a total of 25 drops
central pallor is resistant to (NaCl & water) are in each
lysis because the space can tube.
bloat to extend the capacity, 4. Dispense to each tube one
so the cell can absorb more drop of heparinized or
water without hemolyzing. defibrinated blood. Note:
But a cell without a central Deliver at the same angle
pallor will immediately lyse directly to saline solution.
upon absorption of water like 5. Mix tube and allow to
spherocyte. stand at room temperature
 If the surface area-to-volume or centrifuge for 1 minute at
ratio is still high, the cell is 2,500 rpm.
resistant 6. Examine each tube for
 If the surface area-to-volume initial/incomplete and
ratio is decrease, the cell is complete hemolysis,
fragile  easily lyse  DETERMINE THE
 Cells with decreased surface CONCENTRATION OF NaCl
are-to-volume ratio have a SOLUTION WHERE INITIAL
limited capacity to expand in AND COMPLETE HEMOLYSIS
hypotonic solution and OCCURRED.
therefore lyse even at a less  FORMULA: %NaCl = test tube
hypotonic concentration of number x 0.02
saline than the normal
biconcave cells. o Dacie Method  more accurate
o Functional state of the cell  Hypotonic saline (pH 7.4)
membrane  Buffered at pH 7.4 using PO4
 If the red cell membrane is in order to prevent
defective, it is easily interference of inconsistent
hemolyse. pH
 Like in HS, there is membrane  RECAP: OSMOSIS: movement
problem wherein it lack of fluid in order to attain
integrity due to loss of homeostasis/equillibrium
protein.  Outside is hypotonic, inside is
o Sanford Method  macroscopic isotonic movement of
 Reagent: 0.5% saline + add water from outside to inside
corresponding drops of  Hemolysis: @540 nm
DH2O  INCUBATED OFT: incubate
 SPECIMEN: Heparinized or the sample for at least 24
defibrinated blood hours to detect mild forms of
 PROCEDURE: Prepare HS
different concentration of  It is done because if the
hypotonic saline using the observation is done
following procedure: immediate and the HS is mild
1. Set up 12 test tubes and there will be no hemolysis
label no. 14 -25. seen. false negative result
2. Place in each tube 0.5%  Mild forms of HS: on
NaCl solution (The number of prolonged incubation they
HEMATOLOGY 2

become fully spherocyte


therefore show hemolysis
 PURPOSE: detect mild forms
of HS

2. EOSIN-5’-MALEIMIDE (EMA) BINDING TEST


 Alternative confirmatory test  accurate

3. SODIUM DODECYL SULPHATE-


POLYACRYLAMIDE GEL ELECTROPHORESIS
(SDS-PAGE) VARIANTS:
4. OSMOTIC GRADIENT EKTACYTOMETRY SOUTHEAST ASIAN OVALOCYTOSIS (SAO)
5. ACIDIFIED GLYCEROL LYSIS TEST  In Southeast Asian Ovalocytosis/ (SAO), the
6. AUTOHEMOLYSIS TEST mutation in protein band 3 results to
7. HYPERTONIC CRYOHEMOLYSIS TEST increased membrane rigidity, making the red
8. PCR cells resistant to Plasmodium invasion.
 single-stranded and conformational o Mutation: gene for band 3 increased
polymorphism analysis rigidity (not flexible) of the membrane
 alternative confirmatory test and resistance to invasion by malaria
o BAND 3: gives elasticity and flexibility
of the RBC membrane
NOTE:  Common in southeast asian population
Since HS is a genetic disorder, the confirmatory tests  Since the RBC membrane is rigid and instead
are the genetic studies like chromosomal studies or of showing a pale center area, a band is
polymerase chain reaction (PCR). formed at the center
 PINCER-LIKE CELLS: traverse ridge or
longitudinal slit
2. HEREDITARY ELLIPTOCYTOSIS
 A defect in the skeletal protein interaction HERIDITARY PYROPOIKILOCYTOSIS (HPP)
(spectrin dimmers, ankyrin, protein 3 and 4.1)  Affected red cells are heat sensitive and start
anemia is normocytic normochromic with >25% to bud & fragment at 45-46°C. Normal red
elliptocytes. cells fragment starting at 49°C.
o Proteins involves are horizontal proteins  Budding and fragmentation at 45-46 °C
that support the red cell membrane.  Normal RBC will start to fragment or bud at 49
 Clinical manifestions are mild, or none at all. degree Celsius  vesicle formation and
 Deficiency of glycophorin C fragmentation
 ACQUIRED: <25% elliptocytes of the RBC  But cells in HPP are heat-labile meaning that
even at lower temperature (45-46 C) they
population
easily bud off and fragment
 HEREDITARY: >25% elliptocytes of the RBC
 If they bud off they will form a small vesicle
population which are called microspherocytes
 Splenectomy is not considered buds/fragments from RBC
 PYROPOIKILOCYTES or MICROSPHEROCYTES:
cells that easily buds and fragment @45-46
degree Celsius
 Pyro: means heat/fire cells are resistant to
heat

SPHEROCYTIC HE
 Variant of both HS and HE
HEMATOLOGY 2

 The patient has the abnormal gene resulting  Deficiency will lead to decreased membrane
to spherocytic and elliptocytic HEREDITARY transport mechanism hence it will lead to
 BLOOD SMEAR will show combination of dehydrated or overhydrated cell.
many spherocytes and elliptocytes  Normal cell should maintain more
concentration of sodium outside and
potassium inside but if the defect of the
3. HEREDITARY STOMATOCYTOSIS: transport protein results to:
HYDROCYTOSIS (OVERHYDRATED) & o A NET INCREASE in cellular cations (
HEREDITARY XEROCYTOSIS (DEHYDRATED)  Na and K inside)  cell becomes
 A mild hemolytic anemia characterized by too concentrated which will drive the
increased permeability and flux of both Na+& K+. water from the outside to shift inside
Intercellular water is influenced by ions,  cells gains more water appearing
primarily sodium, thus, a net increase or as STOMATOCYTE: a swollen cell/fully
decrease in cellular cations will influence the hydrated cell
shift of fluid shift into or out of the cell. o A NET DECREASE in cellular cation
almost all of the cations have left the
 ALTERED MEMBRANE TRANSPORT PROTEINS
RBC as the cations leaves the RBC,
 In phospholipid bilayer aside from cytoskeleton, even intracellular water will also shift
peripheral proteins at the inner leaflet or lining, outside the RBC  cells become
there are also proteins that traverse the bilayer dehydrated or dessicated 
which are called integral proteins, which serve as appearing as DESSICOCYTE or
ion channels or transport proteins. Integral XEROCYTE: a dehydrated cell
proteins can also be defective altering the
membrane transport mechanism which can
The blood antigens are
result into:
found on the red cell
o Overhydrated Hereditary
surface like the Rh
Stomatocytosis/Hydrocytosis antigens which add in
 Stomatocytes (hydrocytes) are the integrity of the RBC
formed due to the shift of fluid membrane. Hence their
into the cell (swollen cell) these absence (complete
cells have decreased surface-to- absence: Rh null disease)
volume ratio make the RBC
o Dehydrated Hereditary membrane incomplete
Stomatocytosis/Xerocytosis therefore it is
 Xerocytes (dessicocytes) are compromised.
formed due to the shift of fluid Net INCREASE in cellular
out of the cell (dehydrated cell). cations Decrease
surface-to-volume ratio
These cells have increased
OFT: Increase  prone
surface-to-volume ratio.
to lysis MCV: Decreased
CAUSES: Seen in Rh null
1. Mutation in the RHAG gene Net DECREASE in cellular
 RH Blood Group system: any of the antigen cations Increased
DCcEe surface-to-volume ratio
OFT: Decrease 
2. Absence of stomatin (band 7 region) resistant to lysis
3. Mutations in the PIEZO1 gene (Piezo-type MCHC: Elevated  cells
mechanosensitive ion channel component 1 hemoglobin appears
protein)
HEMATOLOGY 2

concentrated due to 3. Rapoport-Luebering Pathway


intracellular water loss  Production of 2,3 DPG
Characterized by Mild  Regulates hemoglobin affinity to oxygen
hemolytic anemia  NOT involved in ATP production or RBC
hemoglobin protection
SUMMARY: MEMBRANE DAMAGE 4. Methemoglobin Reductase Pathway
HEREDITARY SPHEROCYTOSIS: most affected protein  Maintains hemoglobin in the reduced form
structure are the vertical ones like ankyrin and band 3

HEREDITARY ELLIPTOCYTOSIS: the proteins involved


can be the same with HS but the position of the
protein is horizontal like actin

HEREDITARY STOMATOCYTOSIS: whether in


dehydrated or overhydrated, the problem are the
transport proteins

1. GLUCOSE-6-PHOSPHATE DEHYDROGENASE
DEFICIENCY ANEMIA
 This is the most common RBC enzymopathy
associated with hemolytic anemia, inherited as
a sex-linked disorder. Red cells need G6PD to
generate NADPH and reduced glutathione
HEMOLYTIC ANEMIA DUE TO METABOLIC
(GSH). These reducing potentials are important
ABNORMALITIES (ENZYMOPATHIES)
in protecting hemoglobin from oxidation.
 G-6-PD enzyme is found in hexose
Metabolism Pathway in RBCs: monophosphate shunt which functions to
provide the reducing potential of the RBC or to
1. Embden-meyerhoff Pathway: anaerobic provide the antioxidant property of RBCs.
glycolysis  In the absence of G6PD, hemoglobin is not
 Provides 90-95% ATP protected from oxidation by peroxides. It will be
 Mature RBCs lack mitochondria thus they easily oxidized becoming
produce ATP through EMP
methemoglobin/hemoglobin  iron is in the
ferric state
2. Hexose Monophosphate Shunt or Pentose
Phosphate Pathway:  Oxidized hemoglobin easily denatures and
 Oxidative pathway precipitates as Heinz bodies.  located at the
 Provides 5-10% ATP periphery
 Production of reduced glutathione  main o Heinz bodies are found at the periphery
function attach to red cell membrane inducing
 Glutathione is the main antioxidant property cell rigidity and fragility.
of RBC  detoxifying agent o When RBC with Heinz bodies enters a
microcirculation(splenic sinusoids), a
HEMATOLOGY 2

part of the cell which is flexible can pass


through however the part where there
is inclusion (rigid) will not and will be
detached from the RBC  the
remaining cell will be knit off producing
now a pitted red cells called bite cell,
horned cells, thorned cell or
degmacyte.
 Increased oxidation may be associated with
exposure to oxidant drugs, diabetic acidosis &
during neonatal period.
o Included in newborn screening in the
Philippines.
 G6PD Mediterranean is occasionally associated
with severe hemolytics episodes ( CLASS ENZYMATIC ACTIVITY DEGREE OF
ASSOCIATED
hemoglobin) of following consumption of fava
HEMOLYSIS
beans (favism)
I Severely deficient  Chronic hemolysis
o G6PD Mediterranean - favisms most severe
 PBS: Heinz bodies, bite cells, polychromasia II Severely deficient Acute, episodic
MAIN REACTION: Glucose-6-Phosphate (<10% residual
Dehydrogenase (cofactor NADP; when functions it activity)
becomes reduced into NADPH  reducing potential) III Moderately to mildly Acute, episodic
catalyzes the conversion of Glucose-6-Phosphate into deficient (10% to 60%
6-phosphogluconate. residual activity)
IV Mildly deficient to Absent
Importance of reduced NADP  NADPH normal (60% to 150%)
1. Protects the hemoglobin from being oxidized; the V Increased (>150%) Absent
H of NADPH will be given to hemoglobin reverse the Not all G6PD deficiency is characterized by severe
oxidation and reduces methemoglobin (oxidized hgb) hemolysis, it depends on the type (5). With hemolysis
back to reduced hemoglobin. (1-3); without hemolysis (4-5)
2. Reduce another reducing potential: Reduced
Glutathione
DIAGNOSTIC LABORATORY TESTS FOR G6PD
 Good reducing potential if it is in reduced
DEFICIENCY:
form; it can reduce back methemoglobin into
1. FLUORESCENT SPOT TEST (REDUCTION)
normal form.
 Recommended screening test): hemolysate +
 Major antioxidant property: breaks down
reagent  NADH – NAD (NADH conversion to
hydrogen peroxide (toxic) into water and
NAD results to loss of fluorescence)
oxygen; detoxify peroxides and free radicals
 Blood + Reagent: G6P + NADP  NADPH + 6-
PG
Methemoglobin production: 3.5 himeglobin daily
 RESULTS:
If accumulated can suffer from methemoglobinemia
o With fluorescence: conversion of
 abnormal hgb that can’t transport oxygen
NADP into NADPH
o Without fluorescence: absence of
NADPH; conversion of NADPH into
NADP  reaction does not occur
normally

2. ASCORBATE CYANIDE TEST


HEMATOLOGY 2

 Measures the ability of RBCs to detoxify 


Measures the spontaneous lysis of RBCs
hydrogen peroxide when incubated with when incubated at 37°C for 48 hours.
ascorbate. A brown color develops Depletion of Glucose and ATP during
(methemoglobin) in the absence of adequate incubation results to membrane loss,
RBC G6PD. spherocyte formation, and then ultimately,
 If the patient RBC has a normal G6PD levels, lysis. If positive, test is repeated with addition
it is able to produce NADPH which will cause of Glucose and ATP to protect against
the reduction of glutathione (major autohemolysis.
antioxidant property of red cell that will INTERPRETATIONS:
break down hydrogen peroxide)  Normal sample: 0.2-2% hemolysis; 0%-0.8% /
 DETOXIFICATION OF HYDROGEN PEROXIDE 0.9% if with ATP or glucose
(H2O2)  G-6-PD deficiency: moderate high
 PROCEDURE: autohemolysis, partially corrected by glucose
o RBC (hemoglobin) + ascorbate  PK deficiency: greatly high autohemolysis,
cyanide reagent –incubated--  corrected by ATP
produces H2O2 (hydrogen peroxide,  Hereditary spherocytosis: Greatly high
a toxic radical that destroys hemolysis corrected by glucose or ATP but not
hemoglobin) to a normal value.
o RBC (Normal – G6PD) + H2O2 
Hemoglobin (normal)  pink
solution: cell is maintained in normal 2. PYRUVATE KINASE DEFICIENCY ANEMIA
state  Pyruvate kinase enzyme is found in EMPathway
o RBC ( Decrease – G6PD) + H2O2  (anaerobic glycolysis) which is important in the
Hemiglobin/Methemoglobin  synthesis of ATP. Hence, its deficiency will lead
brown solution: G6PD deficient cell as well to ATP deficiency.
 PK catalyzes phosphoenolpyruvate to pyruvate.
3. METHEMOGLOBIN REDUCTASE TEST
 ATP energizes the Na-K pump which is
 INDIRECT TEST that determines the ability of
necessary in the maintenance of RBC
red cell to reduce methemoglobin back to
membrane
normal hemoglobin.
 All hemoglobin compounds  PK deficiency results to impaired Na+-K+ pump.
(abnormal/normal) are reversible EXCEPT Water and K+ are lost resulting to shrinkage,
sulfhemoglobin. distortion, speculation and premature RBC
 Hemiglobin is reversible. destruction.
 Detects NADPH generation by G6PD. The  RBC morphology is generally normocytic-
transfer of H+ from NADPH to normochromic, with presence of
methemoglobin results to its reduction back polychromasia.
to normal hemoglobin.
o If patient RBC have normal levels of NOTE:
G6PD, then it is able to generate In embden meyerhoff pathway, there are two
enough NADPH (reducing potential) reaction that are ATP producing:
o Methylene Blue can be added to (1) Conversion of 1,3 – biphosphoglycerate into 3-
make the transfer of hydrogen to phosphoglycerate
methemoglobin faster. (2) Conversion of phosphoenolpyruvate (PEP)
o It facilitates a fast transfer of into pyruvate 
hydrogen catalyzed by pyruvate kinase

4. AUTOHEMOLYSIS TEST (SCREENING TEST There is two (2) glyceraldehyde-3-phosphate so there


FOR PK AND G-6-PD DEFICIENCIES) will be 4 ATP molecules produced. However, if there
is a deficient enzyme like PK, ATP production will also
be deficient.
HEMATOLOGY 2

o Without fluorescence: Normal PK:


conversion of NADH into NAD+
(oxidized form)
 If patient RBC have PK, it will
cause the conversion of the
above reaction
 Loss of fluorescence means
normal pyruvate kinase
enzyme

2. AUTOHEMOLYSIS TEST (Screening Test for


IMPORTANCE OF ATP IN RBC: PK and G-6-PD Deficiency)
1. Active Transport: Transport protein + ATP  For diagnosis of PK, G6PD, HS
 The transport of ion is mostly active thus  Incubated in sterile condition at 37 degree
ATP is needed like in Calcium pump and Celsius for 48 hours (2 days)
NaK ATPase pump
 Measures the spontaneous lysis of RBCs
 Facilitated transport: only transport protein when incubated at 37oC for 48 hours.
is needed Depletion of Glucose and ATP during
Thus the effect of ATP deficiency is that the transport incubation results to membrane loss,
of ions along cell membrane becomes abnormal thus spherocyte formation, then ultimately, lysis.
the cell loses more potassium and losses more water If positive, test is repeated with addition of
 crenated/distorted cell that is prone to Glucose and ATP to protect against
lysis/destruction autohemolysis.
 INTERPRETATION:
o POSITIVE RESULT: Hemolysis
o 1st test: positive for hemolysis 2nd
test: add Glucose or ATP repeat
autohemolysis test
 1st test: Greatly
High/Increase Hemolysis;
2nd test: hemolysis is
corrected by ATP  PK
deficiency
 1st test: Moderate Hemolysis
partially; 2nd test: hemolysis
DIAGNOSTIC LABORATORY TESTS FOR PK is partially corrected by
DEFICIENCY glucose  G6PD deficiency
1. FLUOERSCENT SPOT TEST (OXIDATION)  3rd test: Greatly Hemolysis;
 Blood RBC (Hemolysate) + reagent (PEP, 2nd test: hemolysis is
NADH, LD) –incubate--  NADH  NAD+ partially corrected by ATP
o PEP – PK  Pyruvate –Lactate and Glucose  HS
Dehydrogenase (with cofactor NADH) INTERPRETATIONS:
 Lactate  Normal sample: 0.2-2% hemolysis; 0%-0.8% /
 Pyruvate: aerobic product 0.9% if with ATP or glucose
 Lactate: anaerobic product  G-6-PD deficiency: moderate high
 RESULTS: autohemolysis, partially corrected by glucose
o With fluorescence:  PK deficiency: greatly high autohemolysis,
Abnormal/Deficient PK: Presence of corrected by ATP
NADH (reduced form)
HEMATOLOGY 2

 Hereditary spherocytosis: Greatly high that in the presence of antibodies may


hemolysis corrected by glucose or ATP but mediate hemolysis
not to a normal value.  ETIOLOGY:
o HEMOGLOBINURIA: presence of
hemoglobin in the urine due to
ACQUIRED INTRACORPUSCULAR HEMOLYTIC
MECHANISM hemolytic anemia (intravascular
1. PAROXYSMAL NOCTURNAL HEMOGLOBINURA hemolysis)  hemoglobin is directly
(PNH) freed into the plasma and plasma is
 Acquired intrinsic defect which arises in the filtered by the kidney hence will appear
bone marrow stem cells affecting RBCs, WBCs in the urine
and platelets.  Hemolysis mainly is the cause
o The cause of mutation is not yet for hemoglobinuria
established because many factors are o NOCTURNAL: means NIGHT.
involved.  The intravascular hemolysis and
o Acquired but genetic in hemoglobinuria occurs chiefly
nature/hereditary disease at NIGHT  plasma is more
o EFFECT OF MUTATION: the mutation acidified thus the RBCs
has lead to deficiency or absence of hemolyse.
complement defense protein like the o PAROXYSMAL: mean INTERVAL
regulatory protein C3 convertase. The  The lysis does not occur
complement system is part of the everytime or constantly
body’s defense mechanism thus it anytime but it occurs at an
function normally if its activation occurs INTERVAL  NIGHT
normally. The complement protein  Decrease or absence
causes lysis but RBCs are resistant to it  Most cases of PNH, the patient conditions are
because it is protected by C3 related to aplastic anemia indicating that the
convertase however in this disease, the mutation is caused by weaken bone marrow.
RBC does not elaborate these defense o In aplastic anemia, bone marrow is
proteins thus prone to hemolysis. defective, it has decreased function due
Another cause is the deficiency or to infiltration of marrow fats hence it is
absence of GPI which supposedly prone to mutation.
should function to linked complement NOTE:
proteins thus deficiency will lead to The complement system has 3 pathway: classical,
hemolysis. alternative, and lectin pathway in which involve the
 ↓ / (-): complement defense assembly of C3 convertase that will later cleave C3 into
proteins (C3 convertase) C3a and C3b and other fragments. C3 convertase is
 ↓ / (-): Glycosylphosphatidyl injurious to RBC, that C3 may bind with the RBC and
Inositol (GPI)  linked protein stress it. Sooner or later, the RBC bound with
 Affected cells show an abnormal sensitivity to complement hemolyse. However, if the RBC have on
complement in acidified plasma or low ionic their surface the expose CD55 and CD59 which are
strength environment, resulting to protective proteins, the RBC will be protected against
complement. CD55 and CD59 inhihits the further
complement-mediated lysis.
activation of complement.
o Affected cells (acidified plasma/Low
Ionic Strength)  COMPLEMENT- In PNH, CD55 (also called DAF) and CD59 are
mediated lysis deficient/absent, thus on continued activation of the
o COMPLEMENT: a thermo-labile protein complement pathway, there will be formation of
coming from complement activation membrane-attack complex which can also assemble
HEMATOLOGY 2

on surface of RBC together with the complement  PROCEDURE: incubate RBC with the
proteins that will induce further lysis. following:
o Weak acid: complement activation
MAIN MUTATION in PNH occurs in the gene and cause the complement to attach
responsible in the production of the normal GPI  on RBC surface causing stressful
mutation of GPI environment and later hemolysis
GPI: links regulatory protein like CD55 and CD59 on o RBC + N. serum + 0.1% HCl  weak
RBC surface DEFICIENCY OF EITHER/BOTH will lead to acid  POSITIVE
prone complement-mediated lysis. o RBC + Pxt. Serum
o RBC + Inactivated serum  NO
HEMOLYSIS: complement is there
but inactivated; complement
proteins are thermo-labile thus when
heated at 56 degree Celsius will
cause its inactivation.
 Affected RBCs are abnormally sensitive to
complement-mediated lysis in acidified
serum (use of weak acid as reagent)
 Positive screening + Positive confirmatory
test  PNH
 Also test CDA II (HEMPAS)

Currently, flow cytometry procedures are available


that can test white cells for the presence or absence
of GPI-linked proteins. White cells can be examined
DIAGNOSTIC LABORATORY TEST FOR PNH
for reactivity to anti-CD48, CD55, and CD59, all of
1. SUGAR WATER SCREENING TEST which are anchored proteins. A new diagnostic
 Whole blood and normal control are procedure, FLAER (fluorescent- labeled aerolysin) has
incubated at 37 degrees Celsius in a LISS. proved effective in detecting smaller populations of
Hemolysis in the test/sample may indicate abnormal leukocytes in PNH.
complement-mediated lysis. OTHER DIAGNOSTIC TESTS FOR PNH
o LISS promotes binding of
FLOW CYTOMETRY
complement to RBC  straining;
 Able to detect which specific GPI or
while the complement is on the RBC
defense/regulatory protein is deficient or
surface, it stresses the cell until such
defective in affected cells because it made
time the RBC can no longer bear the
use of specific anti- sera
stress and hemolyse
 ANTI-SERA for: CD48, CD55, CD59, WBC and
o WB in LISS at 37°C
other blood cells
o NC in LISS at 37°C
o When blood cell react to the anti-
 RESULTS:
sera means that they possess the
o WB: with hemolysis; NC: no
following markers have normal
hemolysis  valid but should not yet
regulatory proteins
reported  proceed to another test
 GPI – linked proteins; -WBCs reactivity to
 confirmatory test like sucrose
anti-CD48, CD55 and CD59
hemoysis test and Ham’s acidified
serum test
FLAER (FLUORESCENT-LABELED AEROLYSIN)
2. SUCROSE HEMOLYSIS TEST
 It uses fluorescent dyes and has an
 Confirmatory Test
advantage of being able to detect even the
small population of affected cells
3. HAM’S ACIDIFIED SERUM TEST
 More sensitive
 Confirmatory test for PNH
HEMATOLOGY 2

- Caused by pathologic antibodies (IgM


antibodies against the I antigen) that react
ACQUIRED IMMUNE ANEMIC OF INCREASED most effectively at cold temperature (0-10
DESTRUCTION (Antibody Related) degrees Celsius). Often associated with M.
1. ISOIMMUNE HEMOLYTIC ANEMIA pneumoniae & Epstein-barr virus/EBV
 Antibodies comes from another person: infections. Patients exposed to these
o Hemolytic Disease of the Fetus and the infections may develop harmful Anti-I & Anti-
Newborn (HDFN) I antibodies, respectively.
 Rh negative mother; Rh positive
baby IgM Ab  directed against I Ag: 0-10 degree Celsius
o Hemolytic Transfusion Reactions (HTR)  Harmless cold antibodies: Anti-I, Anti-H and
Anti-IH
2. AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA)  Harmful antibodies: produced in infections 
M. pneumonia (Anti-I); EBV (Anti-i) 
 Antibodies produced by patient’s own body
hemolytic anemia
 Happens when the patient’s immune system
 Reynaud’s phenomenon/acrocyanosis:
becomes altered to cause the production of
Cyanotic appearance: red- blue-black
antibodies that will attack the patient’s own coloration plus painful sensation on the
RBC causing lysis. exposed parts of the body due to
 Can be drug-induced, infections, unknown vasooclussion caused by agglutination of the
 WARM AIHA: IgG production reacts @37 RBC by the abnormal antibody.
degree Celsius; more common
o Associated/Pre-exist with CLL, wherein The exposed body parts like ears and fingertips which
the affected lymphocytes are the are the coldest or easily cold part of the body. The
dormant B-cells but they start IgM which is the Anti-I will bind to I antigen causing
producing abnormal antibodies that will now agglutination. So the blood vessels supplying the
distal tissues will be obstruction resulting to non-
attack patient’s own red blood cell
oxygenation  cyanotic  may even lead to
causing WARM AIHA
necrosis/death of tissue.
 COLD AIHA: IgM production reacts @colder
temperature (0-10 degree Celsius)
o Bind with RBCs at cold temperature,
since IgM is pentamer, more RBC can
bind thus agglutinates more.

NOTE:
WARM-ANTIBODY AIHA
- Occurs when the patient’s own immune
system produces anti-RBC antibodies (IgG
3. PAROXYSMAL COLD HEMOGLOBINURIA (PCH)
antibodies) that react most effectively at
warm temperatures  The hemolysis does not occur anytime but
occurs in an interval  during cold temperature
o Technically, hemolysis occurs in warm
temperature but it is called cold because
antibodies bind in this temperature.
 Caused by the binding of the Donath-Landsteiner
antibody to the patient’s red cells following
exposure to cold. Donath-Landsteiner is an Ab
COLD-ANTIBODY AIHA specific for Pp blood group system. It is biphasic
that reacts at cold and warm temperature. At
HEMATOLOGY 2

cold temperature, it binds to the red cell  Caused by abnormal deposition of fibrin or
membrane without causing lysis. Intravascular atheroma, or anatomic defects of the small
hemolysis and hemoglobinuria occurs as the blood vessels resulting to reduced blood vessel
temperature returns to normal body temp. lumen and increased blood pressure. The
 It is immune because of the presence of combination of this abnormal increase in the
antibody: Donath-Landsteiner (D-L) antibody is force of moving cells and the smaller vessel
directed for the Pp blood group (hence, also cause injury and fragmentation of RBCs.
called  anti-P). IgG antibody type but biphasic  When the cell passage is small and is obstructed,
which reacts in both warm and cold the circulation becomes limited and to
temperature. compensate for the decreased blood supply,
o In cold temperature, it does not cause there will be increase in cardiac output and the
hemolysis, it only binds to surface of the effect is increased blood pressure.
RBC. This binding will cause the partial o The combination of increased pressure
activation of complement mechanism pushing the red cell and the limited
from C1 to C4. Nothing will happen if the circulation where the red cell should
patient is maintained in the cold pass through  trauma to red cells
temperature however when exposed to  PBS: presence of schistocytes  hallmark of
warmer/normal (37 degree Celsius) hemolytic anemia
temperature, the D-L antibody will be  Affects or occurs in the small blood vessels
detached and the complement wherein it further narrows due to the:
mechanism will completely (C3-C9) be o Presence of fibrin deposits causing
activated thus hemolysis will occur. obstruction
o Complement – mediated lysis by D-L o Presence of atheroma plaques like in
antibody diabetes mellitus or
 D-L TEST hypercholesterolemia  thickening and
o PROBLEM: presence of antibody which is hardening of blood vessels (rough)
found in the serum o Anatomic defects: blood vessel is very
o TEST: Pxt serum + Reagent cells: O and small  hemolysis
P+( for anti-P) normal cells incubate @
4 degree Celsius transfer to 37 degree 2. DISSEMINATED INTRAVASCULAR
Celsius incubation COAGULATION (DIC)
o RESULTS:  SECONDARY HEMOSTATIC
 HEMOLYSIS: Presence of D-L DEFECTS/COAGULATION DISORDER
antibody  Caused by extensive damage to vessel
 NO HEMOLYSIS: absence of D-L endothelium or exposure to compounds that
antibody initiate clotting resulting to fibrin deposition
along and across the vessel lumen. RBCs are
ACQUIRED EXTRACORPUSCULAR HEMOLYTIC
ANEMIAS fragmented as they are pushed through the
 Vascular OBSTRUCTION caused by fibron clots or vessel by the action of rapid circulation.
threads (DIC and MAHA) and by abnormal  Affects all types of blood vessels: small,
platelet aggregates (TTP and HUS) medium, large and may also affects either
male/female
1. MICROANGIOPATHIC HEMOLYTIC ANEMIA  Test: Coagulation & Fibrinolysis tests
(MAHA)  Due to release the release of thromboplastin-
 SECONDARY HEMOSTATIC like or tissue-factor like substance, it will cause
DEFECTS/COAGULATION DISORDER the activation of both coagulation and
fibrinolysis, the effect of which is because of the
HEMATOLOGY 2

continous activation of coagulation decreased  Shiga toxin attach to glomerular


coagulation factors; Increased activation of capillaries(renal damage)
fibrinolysis will lead to increased fibrin-split causing the release and
product (FSP) thus will result to bleeding activation of vWF which causes
o Decreased coagulation and Increased the platelets to adhere with
FSP them and aggregate 
OBSTRUCTION
3. THROMBOTIC THROBOCYTOPENIC PURPURA o NON-DIARRHEAL: in infant and children
(TTP) / MOSCHKOVITZ SYNDROME following vaccination
 PRIMARY HEMOSTATIC DEFECTS
 Associated with defective ADAMTS-13 which is 5. HEMOLYSIS DUE TO MECHANICAL INJURY
an enzyme that cleaves the ultralong and  Damage to the heart of aorta – RBCs fragment
ultralarge von Willebrand factor so they as they come in contact with damaged heart
become a smaller or medium sized vWF that is valves.
more effective in hemostasis. However the o Patients with artificial or prosthetic
deficiency/absence in ADAMTS-13, the vWF will heart valves
persist as large which easily binds with  Stress or March hemoglobinuria – lysis is caused
platelets. Ultralarge vWF will bind with platelets by mechanical pressure during exercise or
causing them to aggregate on them thus during marching
blood circulation, the platelet aggregates can o Also called Cadet’s,Orthostatic
detached and flow along the circulation and can hemoglobinuria/proteinuria, Cyclic
be deposited elsewhere causing now hemoglobinuria/proteinuria occurs
obstruction.  thrombosis (thrombus) red cell during the day
trauma o Hemoglobinuria: presence of
 Caused by formation of platelet aggregates (due hemoglobin in the urine due to
to activation) resulting to vascular occlusions hemolysis which is caused by stress and
and RBC fragmentation. More common in force against renal vein
young adults and is characterized by fever,  Infections & Infestations
petechiae, neurologic signs and renal disease. o PLASMODIUM (hemolytic condition;
intraerythrocytic parasite (humans),
4. HEMOLYTIC UREMIC SYNDROME (HUS) Babesia (ring forms; intraerythrocytic
 PRIMARY HEMOSTATIC DEFECTS cattles  humans)), Bartonella
 Etiology is not clear, though some cases are bacilliformis (Oroya fever), Clostridium
associated with mild febrile illnesses, certain perfringens, Infectious Mononucleos
immunizations, and gastrointestinal (EBV).
disturbances. o INFESTATIONS: spider bites 
 This is common among infants and young Parasitism outside the body
children manifested with acute hemolysis, renal  Exposure to Chemicals and Toxins
failure, neurologic signs, abdominal pain,  Thermal Injury
vomiting, and diarrhea.
 Associated with either diarrheal and non-
diarrheal condition
o DIARRHEAL: seen commonly in young
individuals caused by shiga
(Shigella/Salmonella) or verotoxin
producing organism like E.coli O15H:7
HEMATOLOGY 2

BLOOD LOSS ANEMIA (POST-HEMORRHAGIC


ANEMIA)

NOTE:
Post-Hemorrhagic because anemia occurs after the
blood loss.
TYPES:
1. ACUTE
 The amount loss is >20% of patient’s blood
volume and the occurrence is too short like in
overt bleeding (gunshot wounds, accident,
trauma)
2. CHRONIC
 The amount of blood being is loss quite low
but on prolonged duration like GIT bleeding,
Menstruation, Repeated Pregnancy. Gradual
loss of blood.

ACUTE POSTHEMORRHAGIC ANEMIA CHRONIC POSTHEMORRHAGIC ANEMIA


It is acute when the amount of blood loss is >20% of the Loss of small amount of blood but in long duration. In
patient’s blood volume. chronic blood loss, there is no disruption in the blood
EXAMPLE: Normal blood volume is 5 Liter  20% is 1L volume but hemoglobin is loss therefore even iron is lost
If a patient loss a 1L of blood in a single incident  ACUTE leading to iron deficiency anemia.
BLOOD LOSS  most patient can adjust to a blood loss of Morphology: before becoming a full-blown (Stage 1and 2)
20% however more than 20%  patient may suffer in IDA: NORMOCYTIC-NORMOCHROMIC  Stage 3:
HYPOVOLEMIA (decrease in blood volume) MICROCYTIC
Hematocrit: PRBC/Total Volume

When a patient loses blood, it will not select RBC only but
it is a loss of proportionate amount plasma and RBC. Thus
both plasma and RBC will decrease and when we get the
hematocrit immediately, it will appear normal. But there
will be sudden decrease in platelets and after an hour will
rise. There will be decrease in platelets because they will
be needing in arresting the bleeding by forming clots and
increased due to splenic mobilization which is caused by
stress.

Since there is hypovolemia, there will be vasoconstriction


which will decrease the blood flow however it should not
be remain as that for too long thus the body will adjust to
bring back blood vessel into its normal size. So in order to
HEMATOLOGY 2

have vasodilution, it will cause a shift of fluid from


extravascular and interstitial compartment into the
circulation in order to expand it thus causing an increased
plasma volume over the red blood cell  hemodilution
(will only correct blood volume)  decreased hct and
hgb. Starting 3-5 days after the blood loss, the BM will
compensate by producing more blood cells and releasing
even immature blood cells: red blood cells
(reticulocytes/shift cells) and leukocytes (immature
neutrophil: shilling’s hemogram: shift to the left:
increased in young forms).

BLOOD MORPHOLOGY
Initial: normocytic-normochromic/ unchanged
depending on px blood pic. Later (3rd -5th day): will
become transient macrocytic  immature cells 6th day:
no macrocytosis: immature forms start to mature.

1. ACUTE POSTHERMORRHAGIC ANEMIA o Reticulocyte count and its associated


 Blood loss over a short period of time in corrections can be used to assess
amounts sufficient to cause anemia. Normal bone marrow erythropoietic activity.
individuals are able to compensate for rapid  RETICULOCYTE: 5th stage in the erythopoiesis
blood loss of up to 20% of the circulating blood  first non-nucleated stage but it has
volume with few symptoms. remnant of RNA; fully hemoglobinized
o Young erythrocytes that are in a
 Major manifestations of anemia are due to
discrete, penultimate phase of
depletion of blood volume
maturation where the nucleus has
 RBCs are Normocytic-Normochromic followed been removed, however some of the
by transient Macrocytosis and increased extranuclear RNA remains in the
polychromasia cytoplasm.
o The term reticulocyte is derived from
2. CHRONIC POSTHEMORRHAGIC ANEMIA the fact that the cell contains a small
 Blood loss in small amounts over an extended network of basophilic material called
period of time. This results to gradual iron loss reticulum which is demonstrable only
and slower regeneration of red cells. by supravital stain.
 At first, the anemia is Normocytic- o In the peripheral blood they are called
polychromatophilic erythrocytes.
Normochromic and gradually becomes
 PRINCIPLE: Supravital staining of RNA
Microcytic-Hypochromic
o The ribosomal RNA is stained
supravitally. Any non-nucleated RBC
NOTE: that contains stained
RETICULOCYTE COUNT particles/granulofilamentous
 It measure the erythropoietic activity of the materials are counted as reticulocyte.
bone marrow.
HEMATOLOGY 2

o Supravital staining: staining cells RETICULOCYTE POLYCHROMATOPHILI


while in their living state C CELLS
o STAINS: Most commonly used: New STAIN: Supravital stain Without granules 
Methylene Blue and Brilliant Cresyl The granules are arranged RNA is not precipitated
Blue; Others: Janus Green, Nile Blue in “reticulum pattern” Non- but the cell will be seen
sulfate nucleated; with 2 or more as polychromatophilic
 METHOD OF STAINING: Wet method and Dry blue-stained (polychromasia)
Method (schilling’s method) particles/granulofilamento showing a lot of colors
 SPECIMEN NEEDED: EDTA-anticoagulated us  precipitated RNA (grayish, bluish,
blood or capillary blood greenish) due to
 PROCEDURE: mixture of the cell.
a. Mix equal amounts of filtered stain COMPOSITION OF
and EDTA-anticoagulated blood or WRIGHT’S STAIN:
fresh capillary blood in a small test Methylene blue (basic
tube dye) and eosin(acidic
b. Incubate mixture at room dye)  reticulocyte
temperature/using incubator for 10 – have hemoglobin
15 minutes (eosin) and RNA(MB)
o Purpose of incubation: to colors are mixed
promote the staining of RBC.
Since the RBCs are alive, they
resist staining due to intact
membrane.
c. After incubation, mix thoroughly and
prepare a wedge smear.
d. Examine smear using 100x objective
(OIO) using the routine light
microscope method. Select an area NOTE:
where erythrocytes are close but not METHODS OF COUNTING
overlapping and reticulocytes appear 1. LIGHT MICROSCOPE
to be well stained.  Reticulocytes are enumerated among 1000
RBCs in areas where RBCs are close but not
APPEARANCE OF RBCs and RETICULOCYTES: overlapping and reticulocytes appear to be
 MATURE RBCs: light to medium green without well stained.
granules
 RETICULOCYTES: light green with granules/ 2. MILLER DISK METHOD
granulofilamentous that stain deep blue.  Still made use of light microscope but with
additional component attached to one of the
e. Count the number of reticulocytes in oculars (miller disk).
1000 red cells. Reticulocytes should  The calibrated Miller disc appears in the field
also be counted as erythrocytes. with 2 squares: a large square and a small
f. Calculate as follows: square inside the large square which is 1/9 of
𝑵𝒐. 𝒐𝒇 𝒓𝒆𝒕𝒊𝒄𝒖𝒍𝒐𝒄𝒚𝒕𝒆𝒔 × 𝟏𝟎𝟎 the size of the larger square.
% 𝑹𝒆𝒕𝒊𝒄𝒖𝒍𝒐𝒄𝒚𝒕𝒆𝒔 =
𝟏𝟎𝟎𝟎 𝑹𝑩𝑪  Reticulocytes are counted in the large square
while the RBCs are counted in the small
square in successive fields on the film until a
total of 500 RBCs have been counted.
 Counterstain is not advisable
 Reticulocytes have lower specific gravity 
after incubation, the reticulocyte appear to
HEMATOLOGY 2

settle in the upper portion  always mix to NOTE: INCLUSIONS & ARTIFACTS CONFUSED WITH
avoid false decrease count RETICS
 Patient with hyperglycemia: Increase glucose  HOWELL-JOLLY BODIES
 inhibit staining  false decrease - Seen in Romanowky and methylene blue stain
 TWO SQUARES: but reticulocyte (RNA) will not be seen in
o LARGER SQUARE: for reticulocyte romanowsky stain.
counting
o SMALLER SQUARE: for RBC counting  PAPPENHEIMER BODIES
(mature and immature) Size: 1/9 of - Pappenheimer bodies are present in
the large square romanowsky, supravital stain and Prussian
 REFERENCE VALUES: blue stain (Siderotic granules) but reticulocyte
o Adults: 0.5 – 1.5% are negative in Prussian blue.
o Newborn: 2.0-6.0%
 HEMOGLOBIN H INCLUSIONS
- Hemoglobin H have no pattern; just
distributed in the red cell making the cell as
pitted golf ball whereas reticulocyte have
reticulum pattern

 HEINZ BODIES
- Most confusing because Heinz bodies are not
seen in romanowsky like the reticulocyte’s
RNA and are both present in supravital stain.
To differentiate, observe closely. Heinz bodies
are often few and found at the periphery
while reticulocyte RNA follows a reticulum
pattern

 ARTIFACTS
- Dust particles  move the the fine
𝑹𝒆𝒕𝒊𝒄 %
𝑻𝒐𝒕𝒂𝒍 𝒏𝒐. 𝒐𝒇 𝒓𝒆𝒕𝒊𝒄𝒔 𝒊𝒏 𝒍𝒂𝒓𝒈𝒆 𝒔𝒒𝒖𝒂𝒓𝒆 × 𝟏𝟎𝟎 adjustment and if the focus is lost. The
= artifacts will glisten.
𝑻𝒐𝒕𝒂𝒍 𝒏𝒐. 𝒐𝒇 𝑹𝑩𝑪𝒔 𝒊𝒏 𝒔𝒎𝒂𝒍𝒍 𝒔𝒒𝒖𝒂𝒓𝒆 × 𝟗
- OUT OF FOCUS: refractile  artifacts
disappearance  retics
HEMATOLOGY 2

NOTE:
ASSOCIATED CORRECTION:
1. ABSOLUTE RETICULOCYE COUNT HEMOGLOBINOPATHIES
 Actual number of reticulocytes in 1 Liter of  Hemoglobinopathies are results of structural
whole blood defects in the globin component of hemoglobin.
 Formula: The structural defect result from the alteration
𝑅𝑒𝑡𝑖𝑐 % 𝑅𝐵𝐶 𝑐𝑜𝑢𝑛𝑡 (× 1012 /𝐿) × 1000 of the DNA genetic code for the chains leading to
𝐴𝑅𝐶 =
100 the production of abnormal hemoglobins known
as hemoglobin variants.
 Reference value: 25,000 -75,000/uL  NOMENCLATURE:
o Denotes the affected polypeptide chain
2. CORRECTED RETIC COUNT / RETICULOCYTE
o The sequential amino acid number(s)
INDEX
affected (primary structure)
 This corrects the reticulocyte count to a
normal hematocrit to allow correction for the o The helix number involved (secondary
degree of patient anemia. The percentage of structure A-H).
the reticulocytes may appear increased o The nature of the abnormality (amino
because of early release in the circulation or acid substitution, deletion, addition or
because of a decrease in the number of globin chain fusion).
mature RBCs in the circulation.
 Also called Hematocrit Index or Hematocrit
correction
 Formula:  Disorder in structure
𝐻𝑐𝑡 (𝐿/𝐿)
𝐶𝑅𝐶 = 𝑅𝑒𝑡𝑖𝑐 % ×  Results from the alteration of the DNA genetic
0.45 𝐿/𝐿 code for the chains conditions characterized by
 Reference value: 1% (depends on the degree
qualitative structural abnormalities of the globin
of anemia)
polypeptide chains that result from alteration of
3. RETICULOCYTE PRODUCTION INDEX OR the DNA genetic code for those chains. Structural
SHIFT CORRECTION (RPI) abnormalities may involve amino acid
 Index calculated to correct for the presence of STRUCTURAL DEFECTS ON A OR B CHAIN RESULTING
shift reticulocytes that otherwise falsely TO HB VARIANT:
increased the visual reticulocyte count. 1. SUBSTITUTION
 General indicator of the rate of erythrocyte  Ex.: Hb Hb S: β 6(A3) Glu val
production INCREASE above normal in
anemias
2. DELETION
 Formula:
𝐶𝑜𝑟𝑟𝑒𝑐𝑡𝑒𝑑 𝑅𝑒𝑡𝑖𝑐𝑢𝑙𝑜𝑐𝑦𝑡𝑒 𝐶𝑜𝑢𝑛𝑡  some amino acid is deleted
𝑅𝑃𝐼 =  reason of thalassemia
𝑀𝑎𝑡𝑢𝑟𝑎𝑡𝑖𝑜𝑛 𝑡𝑖𝑚𝑒 (𝑑𝑎𝑦𝑠)
 Reference value: 1 if hct is 0.45  Ex.: Hb Gun Hill: (β91 to β 95)0
Patient’s Hct (%) Correction factor/
Maturation time 3. FUSION
(days)  Hb Lepore-Baltimore: (G(1-50) B(86-146)) caused
40-45 1.0 by cross over during mitosis resulting to fusion
35-39 1.5
25-34 2.0 4. ELONGATION
15-24 2.5 5. Hb CONSTANT SPRING (a+31c)
<15 3.0
Clinical Significance of Reticulocytes: Increased in
hemolytic and hemorrhagic anemias
HEMATOLOGY 2

NOTE: CELLULOSE ACETATE ELECTROPHORESIS (PH 8.4-8.6)


REASON FOR DELETION & ELONGATION: mutation in  For the detection&preliminary identification
genetic code of both normal abnormal Hbs particularly, Hbs
REASON OF HEMOGLOBINOPATHIES: mutation of A, F, S & C.
gene  PROCEDURE: a small quantity of red cell
hemolysate is placed on the cellulose acetate
NOMENCLATURE: membrane between the center & the
 COMMON NAME cathode. An electrical field is created in the
 Morphology: sickling Hb (Hb S) chamber through the use of a power supply&
 Place where they were discovered: (Hb the ones with greatest charges migrates
Boston) towards the anode.
 Hemoglobin molecules have a net NEGATIVE
 SCIENTIFIC NAME charge at alkaline pH, & therefore migrates
 Gives structural defect of Hb towards the anode
 Ex: Sickling Hb Hb S: β 6(A3) Glu val  BASIS OF MIGRATION: net Negative charge
 B: affected polypeptide chain because all of them are negative but not all
 6: sequential amino acid number(s) affected have the same number of negative charges
(primary structure). Note: Hb that has the greatest no. of Net
 A3: Helix number involved (secondary charges migrates fastest towards the anode
structure: A-H); not necessary as long as farthest from the point of application but the
primary structure is indicated Glu:nature of Hb with few Negative charges migrates slower
the abnormality (substitution, deletion,  Owing to the variations in amino acid content
addition or globin chain fusion) of different Hbs the net charge of each varies&
 PRIMARY STRUCTURE: sequence or this determines their rate of mobility in the
arrangement of amino acid electrical field.
o Hemoglobinopathies commonly  MAJOR Hb: A (adult), F (fetal), S (most
affected the B chain. common abnormal Hb that causes sickling of
RBC) &C (second most common)
 BANDS: indicates the migration
NOTE:  FAST Hb: any other Hb that migrates faster
 HEMOGLOBIN ELECTROPHORESIS: than A such as H & I
Laboratory test for variant identification and
separation.
o For variant identification
o Follows general principle of
electrophoresis. A – migrates fastest, followed by F
 PRINCIPLE: utilizes an electric chamber that C – slowest followed by S
has both cathode & anode electrons. Arrangement: CSFA
o Hb like proteins are amphoteric.
o Proteins are somewhat amphoteric
wherein its negative charge is
neutralized by the + charge thus it can
show a net Negative & Positive
charges thus it can behave depending
on the pH of the medium.
HEMATOLOGY 2

CITRATE AGAR ELECTROPHORESIS (PH 6-6.2)


 PURPOSE: For the separation of Hbs that
migrate together on cellulose acetate
 Also useful in detecting small amounts of
either HbA or F in the presence of large
amount of others& in revealing small amounts
of adult Hbs A & S present at birth in cord
blood
 PRINCIPLES:
o Samples are place at the center
between cathode & anode & apply
external electricity& migration will
occur
o Hbsare identified by their migration
toward the anode & cathode&
comparing the migration to that of
known control samples
 BASIS: Hbsare separated based on the
interactions among Hb variants, agar & citrate
buffer in addition to the altered electrical
charge of the various Hbs at acid Ph.

S & C migrates alone


F & A migrates towards the cathode
S & C migrates towards the anode
Arrangement: FASC

NOTE:
SUMMARY OF CHARACTERISTICS OF HBF:
 Resistant to both acid elution & alkaline
denaturation.
 In electrophoresis, it is slower than HbA

FUNCTION:
 Has high affinity to O2 thus when it goes to the
tissues, it will not readily release the O2.
 Decreased affinity to 2-3
Diphosphoglycerate: molecule that regulates
affinity of Hb to O2.
HEMATOLOGY 2

CRITERIA CELLULOSE ACETATE ELECTROPHORESIS CITRATE AGAR ELECTROPHORESIS


pH pH 8.0 – 8.6 ( Turgeon) pH 6.0-6.2 (Bishop)
pH 8.4 - 8.6 (Bishop)
Principle A fresh hemolysate made from a packed RBC Citrate agar electrophoresis is performed at an
( Basis/bases of sample is applied to a cellulose acetate plate acid pH after abnormal haemoglobin is detected
hemoglobin using a buffer of alkaline Ph and electrophoresis on cellulose – acetate electrophoresis.
separation) is performed. After electrophoresis, the Hemoglobin F, with the fastest cathodial
membrane is stained and cleared. The patient’s mobility, is also the most soluble, probably
haemoglobin migration is compared with that because it is most resistant to denaturation at pH
of a control test interpretation. A rough 6. Adult hemoglobins with solubility similar to
estimate of proportions of different that of haemoglobin A, such as D, E, G, O, I and so
hemoglobins may be made using a forth, move with haemoglobin A. The relatively
densitometer. The order of electrophoretic insoluble haemoglobin S moves behind
mobility, from slowest to fastest, is haemoglobin A, and the even more insoluble
hemoglobins C, S, F and A. There are several haemoglobin C moves behind haemoglobin S.
mnemonic devices for remembering the
migration pattern, such as accelerated, fast,
slow, and Crawl. Hemoglobin that migrates
beyond haemoglobin A is termed fast
haemoglobin.
Hemoglobin Bart’s and haemoglobin H both
migrate here. Any abnormal haemoglobin or
any normal haemoglobin A2 and F, should be
confirmed. If there is abnormal haemoglobin,
confirm with citrate agar electrophoresis and
solubility test if haemoglobin S is suspected. If
an increased amount of A2 or F occurs, then
quantify the amount.
Hemoglobin D and G comigrate with
haemoglobin S in this method.
Importance/ Hemoglobin electrophoresis by cellulose An important factor in determining the mobility
purpose in acetate is useful in identifying and quantifying of hemoglobin
haemoglobin haemoglobin variants and abnormal quantities is solubility and provides ready separation of Hbs
separation of haemoglobin fractions. It is rapid and that migrate together on cellulose acetate: S
reproducible and separates the major Hb from D and G, and C from E and O.
variants S, D, G, C, and E from Hb A.
Quantification of the major bands is easily
accomplished.

SEVERAL ABNORMALITIES OF HEMOGLOBIN  SIDEROBLASTIC ANEMIA: problem in the


utilization of iron due to deficiency in
1. HEME IS ABNORMAL
protoporphyrin 9
 Iron is deficient thus affecting Hb synthesis
leading to IDA
 Deficient Protoporphyrin IX because of enzyme
2. GLOBIN IS ABNORMAL
defectalong the synthesis leading to excess
 Ex: Hb A1 - 2 A w/ 4 genes & 141 amino acids &
iron&iron will not be utilized in the absence of
2 B chains w/ 2 genes & 146 amino acids
protoporphyrin 9. Unused iron will precipitate
 2 FACTORS:
into siderotic granules leading to sideroblastic
o Alpha Thalassemia
anemia
HEMATOLOGY 2

o B Thalassemia Sickle cells may occlude the marrow


sinusoids, and the lungs, which are the
VARIANTS CAUSED BY B-CHAIN SUBSTITUTION site of frequent infarcts in adults.
- Most common cause of hemoglobinopathies  Infectious crises: Due to abnormal splenic
function and depression of immune
HEMOGLOBIN S (SICKLING HEMOGLOBIN) function, patient becomes susceptible to
infections. In young patients,
 Results from the substitution of glutamic acid by Streptococcus penumoniae appears to be
valine on the 6th position of the B-chain the major infectious agent.
 Inheritance may be homozygous (SS) known as  Splenic sequestration crises: The spleen
sickle cell anemia, or heterozygous (AS) known enlarges and sequesters more blood and
as sickle cell trait. hypovolemia. This may lead to shock and
 Hb S is fully soluble. Sickling occurs when oxygen death
decreases at the tissue level because of the  Aplastic crises: Infection and fever cause
formation of tactoid /fluid crystals. temporary reduction in RBC count, Hb,
Hct and Reticulocytes
 SICKLE CELL ANEMIA
ORGANS AFFECTED BY SICKLE CELL ANEMIA
- Anemia is usually severe;
(mainly related to vaso-occlusion)
characterized by normocytosis
 Liver (hepatomegaly and liver
(normocytic-normochromic)
malfunction)
- Blood Smear shows: Polychromasia;  Heart (Cardiomegaly & heart failure due
Sickle cells, target cells, ovalocytes, to iron deposition)
schitocytes; Howell-Jolly and  Spleen (Splenomegaly, Abnormal splenic
Pappenheimer bodies function & Splenic infarction)
- Growth and sexual maturation lag  Skin (ulcers/scars); Kidneys (infarcts &
behind that of normal adolescents hematuria); and the lungs
- Electrophoresis reveals HbS 80-90%
and Hb F 10-20%
 SICKLE CELL TRAIT
NOTE: - Hb A is present in higher percentage
 Sickle cell crises: Any situation that and compensates for HbS. Patients
produces excessive deoxygenation of usually have no symptoms unless in
RBCs may cause a crisis. Every organ in the cases of extreme tissue hypoxia
body is affected and include pain of many - Electrophoresis: Hb= S 30-45%
types.
 Vaso-occlusive crises  Hb S-THALASSEMIA
o Occur when rigid sickle cells
- Doubly heterozygous condition in
increase blood viscosity leading to
which the mutant genes for both
development of microthrombi &
Hb S and Thalassemia are inherited
microinfarctions in the joints,
extremities and in major organs. by a single individual. Hb S-a is
o “Hand-foot syndrome” or clinically insignificant while Hb S-B
dactylitis refers to painful has clinical features similar to,
swelling of the back of the although not as severe as sickle cell
affected hands and feet due to anemia.
obstruction caused by many
sickle cells. NOTE:
 Bone, Joint, and other crises: Pain occurs Other sickling Hbs: C Harlem; S Travis; C
in the joints where sickle cells accumulate. Zinguinchor
HEMATOLOGY 2

Hemoglobin C (B6 glulys): Second most  Hemoglobin O-Arab (B 121 Glu -> Lys) Rare,
common hemoglobin variant found in Israel, Bulgaria, Egypt, Romania,
Jamaica, Kenya
 Hb G- Philadephia (a 68 Asn->Lys)
 Hb Gun Hill (B 91-95) Leu His Cys Asp Lys -> 0
 Hb C DISEASE (CC)  Hb Constant Spring (CS) a+31c (142 Gln) Due
- Characterized by a mild to moderate to elongation of the a-chain (31 amino acids)
normocytic-normochromic anemia  Hb Lepore-Baltimore( (1-50) B (86-146)) Due
with numerous target cells. Under to fusion of B
low oxygen tension or when  Hb C-Harlem/ Hb C: Georgetown 2 amino
dehydrated, Hb CC tend to acids substitution (B 6 Glu ->Val; Asp ->Asn)
crystallize into hexagonal or rod-  Hb Chesapeak: has increased affinity with
shaped crystals with blunt ends. The oxygen
crystals does not alter the red cell’s  Hb Kansas: has decreased affinity with oxygen
shape but maked the cells rigid.

 Hb TRAIT (AC)
- A heterogygous hemoglobinopathy.
Clinical symptoms are similar with
Hb SS, though milder and with fewer
complications. Often, the red cells
may appear with folded cytoplasm.
Hb SC crystals often appear
fingerlike or with projections.
Electrophoresis reveals equal
amounts of Hb S and C
LABORATORY TESTS FOR UNSTABLE HEMOGLOBIN
NOTE:
1. HEART DENATURATION/HEAT INSTABILITY
 IbM (M-Saskatoon; M-Boston; M-Iwate; M-  Blood sample is incubated at 50 degrees
HydePark; M-Mulwaukee): These are caused celcius for 3 hrs.
by structural defects (amino acid substitution)
in either the a or b chain. The structural defect 2. ISOPROPANOL PRECIPITATION
in the globin chain results to non-protection of
 Blood sample is incubated with 17%
iron from oxidation (methemoglobinemia).
isopropanol at 37°C
Patients appear cyanotic (with lavender-blue
 PRINCIPLE: Normal hemoglobins have intact
skin) due to tissue hypoxia. Babies may also
bonds and remain in the solution when
show clubbed fingers at birth. Blood appears
heated or incubated with Isopropanol.
brown with numerous Heinz bodies.
Unstable hemoglobins have weaker bonds
 Hb Barts (4 y-chains) and H (4-B chains):
and easily denature or precipitate under these
Found in patients with a-Thalassemia
conditions.
 Hb Barts disease: leads to stillbirth and
hypoxia because Hb Bart’s affinity to O2 is 3. HEINZ BODY STAINING
high
 requires supravital staining (e.g. crystal violet)
 Hb H diseases: leads to anemia

OTHER SIGNIFICANT ABNORMAL HEMOGLOBINS:


 Hemoglobin D Hb D-Punjab/Hb D: Los
Angeles (B121 glu->Gln)
 Hemoglobin E (B 26 Glu ->lys)
HEMATOLOGY 2

NOTE: ADDITIONAL INFORMATIONS:


 The HbS GENE, when present in homozygous Hb S (Sicking Hb)
form, is an undesirable mutation, so a  The most well-known hb variant
selective advantage in the heterozygous form characterized by substitution of glutamic acid
must account for its high prevalence and by tRNA to valine on the 6th amino acid
persistence. Malaria is possibly the selecting position of the β- chain.
agent because a concordance exists between  Glutamic: CAG Hb S: β 6(A3) Glu  val
the prevalence of malaria and Hb S. Sickling  Valine: GUG
might protect a person from malaria by either  The effect of alteration from glutamic acid to
(1) accelerating sickling so that parasitized valine is a negative charge wherein glutamic is
cells are removed or (2) making it more highly negatively chargecompared to valine
difficult for the parasite to metabolize or to  thus leading to loss of negative charges& Hb
enter the sickled cell. becomes less negative
 SICKLE CELL TRAIT (AS) confers partial  In electrophoresis in cellulose acetate, S
protection against lethal Plasmodium migrates slowly than A & F due to substitution
falciparum malaria. Multiple mechanisms for of amino acid 6 Inheritance: Homozygous (SS)
this have been proposed, with a recent focus – sickle cell anemia
on aberrant cytoadherence of parasite-  Heterozygous (AS): sickle cell trait
infected red blood cells (RBCs). Here we  EFFECTS OF HB S:
investigate the mechanistic basis of AS o Blacks who are suffering from this are
protection through detailed temporal protected from being infected from
mapping. We find that parasites in AS RBCs developing severe falciparum malaria
maintained at low oxygen concentrations stall (Resistance). Hb C, E, B-Thalassemia
at a specific stage in the middle of intracellular &G-6PD deficiency also confer
growth before DNA replication. We protection in terms of resistance but
demonstrate that polymerization of sickle not immunity meaning these patients
hemoglobin (HbS) is responsible for this may still be infected but may not
growth arrest of intraerythrocytic P. proceed to chronic stage.
falciparum parasites, with normal hemoglobin o Falciparum are intracellular species&
digestion and growth restored in the presence when present in RBC& when RBC is
of carbon monoxide, a gaseous antisickling sickle the parasite inside also dies
agent. Modeling of growth inhibition and does no complication arise
sequestration revealed that HbS o Falciparum metabolizes Hb, when Hb
polymerization-induced growth inhibition crystalizes it forms into a crystal&
following cytoadherence is the critical driver falciparum may no longer metabolize
of the reduced parasite densities observed in the Hb
malaria infections of individuals with AS. We o RBC w/ falciparum will be
conclude that the protective effect of AS phagocytosis before it causes severity
derives largely from effective sequestration of
infected RBCs into the hypoxic HOMOZYGOUS (SS) SICKLE CELL ANEMIA
microcirculation.  Severe anemia because the patient does not
synthesize the normal Hb A
 On electrophoresis:
o Hb S: 80 -90%
o Hb F: 10 -20 %
o Hb A2: Normal
 Blood Smear: Polychromasia (Reticulocytes
are increased) ; Sickle cells, target cells,
ovalocytes, schistocytes ; Howell-Jolly &
Pappenheimer bodies
HEMATOLOGY 2

 Increased RDW
 Retarded growth&sexual maturation&patient Hb S – THALASSEMIA
suffers sickle cell crises Main cause of Sickle  Hb S-A
Cell Crises: sickling  Hb S-B – more severe
 Normally when Hb S is fully oxygenated, Hb S
is fully soluble OTHER SICKLING HBS:
 But when Hb S is w/ reduced O2, Hb S  Abnormality: Hb S: β 6(A3) Glu val
+ unique B-
becomes insoluble wherein it polymerizes substitution
(change in molecular arrangement) &forms  Hb C-Harlem, C-Ziguinchor, S-Travis
into tactoid crystals/fluid crystals (elongated
& thin, pointed at both ends) leading to
sickling &rigid cell membrane thus it is no NOTE:
longer adjustable leading to vassoocclusion HEMOGLOBIN C
thus it cannot easily pass through the small  A beta structural defect, where glutamic acid
circulations is substituted w/ lysine
 Sickling is reversible but repeated sickling  Composition: B 6 (A3) Glu Lys
leads to permanent sickling due to permanent
damage to RBC membrane HETEROZYGOUS: Hb C Trait (AC)
 Causes vasso-obstruction  RBCs – slightly hypochromic
 VASOOCCLUSIVE crises occurs commonly on  Less abnormalities
small circulations like in the fingers &toes thus
hand-foot syndrome / dactylitis (inflamed or HEMOGLOBIN SC DISEASE
swollen, cyanotic due to absence of  Signs & symptoms are milder than SS but
circulation) occurs more severe than sickle cell
 VASSOOCCLUSION also occurs in bone&joint  Electrophoresis: C = S = 50 – 50%
causing pain  Cytoplasm appears folded like a pocketbook
 SPLENIC CIRCULATION is also minute, since cells (RBC w/ cytoplasm folded)
spleen also serves as a filter of blood thus its  Hb SC easily crystalize forming a fingerlike
circulation is too small, that only deformable projection w/ more than 1 protrusions where
red cell may pass through. Due to increase one is long&protrude away. It also in the form
activity of spleen&vassoocclusion, spleen also of hand in gloves crystals or pointing – finger
enlarges (splenomegaly) causing or Washington monument crystal
sequestration of more blood leading to
hypovolemic shock. When spleen is
enlarged&bone marrow is affected thus both OTHER ABNORMAL HEMOGLOBINS
are abnormal in function leading to decrease 1. Hb METHEMOGLOBIN
blood production to the marrow (aplastic  M-Saskatoon, Boston, Iwate, HydePark,
crises) thus the immune system is defective & Milwaukee = all have different amino acid
the patient becomes prone to infection substitution but the effect is the same &not
(infectious crises) as a result of all of this organ protected from oxidation
damage occur (marrow lungs)  Methemoglobinemia w/ congenital cyanosis
 The no. 1 cause of death is infection& in due to overwhelming oxidation even at baby
children the most common is S. pneumoniae stage& the blood appears chocolate brown
 Easily precipitate&forms into Heinz bodies
SICKLE CELL TRAIT (AS)
 Predominant Hb is A, only 30-45% is Hb S 2. Hbs WITH ABNORMAL AFFINITY TO OXYGEN
 Patient usually have no symptoms&sickling is a. Hb w/ Increase Affinity
uncommon to occurunless in cases of extreme  Ex.: Hb Chesapeake (a 93 Arg Leu) –
tissue hypoxia because S is minimal in affects affinity of Hb to O2 &O2
 concentration will shift to left
HEMATOLOGY 2

b. Hb with Decrease Affinity 1. SEALED WHOLE BLOOD METHOD (SCRIVER


 Ex.: Hb Kansas (B 120 Asp Thr
)– & WAUGH)
shift to the right  PRINCIPLE: RBC take on a SICKLE-LIKE SHAPE
when oxygen supply to the red cell is
NOTE: Abnormal Hb are connected by weak bonds decreased. HbSforms INSOLUBLE TACTOID
thus they easily precipitate & flocculate. CRYSTALS when exposed to LOW OXYGEN
TENSION.
 PROCESS: Perform IN VIVO. Using a RUBBER
TEST FOR UNSTABLE HB BAND, the rubber band is TIED at the BASE of
1. ISOPROPANOL PRECIPITATION TEST the FINGER or at the BALL of the finger
 When heated, 17% Isopropanol at 37 C intended to be prick& maintain it for at least
normal Hb do not precipitate but few will 5-10 minutes to cause obstruction in order to
do&unstable Hb will easily precipitate. obstruct the circulation thus decreasing the
 Purpose of Isopropanol: weaken the bonds oxygen& Hb S will form into crystals. Prick
the deoxygenated finger & drop into the
2. HEAT DENATURATION/INSTABILITY TEST center of the slide &cover w/ cover slip. Seal
 At 50°C for 3 hours, normal Hb will not the sides w/ petroleum jelly or paraffin to
flocculate but unstable Hb will prevent entry of atmospheric O2 which
contains abundant O2 which may cause the
3. HEINZ BODY STAINING sickled cell to revert to normal shape.
 If ppt & flocculation happens in vivo forming Incubate for 1 hour at room temperature
Heinz bodies which are not easily seen in &examine microscopically if still none,
normal wrights blood smear incubate for 2 hours until 3rd hour. Perform
 Supravital staining is employed (brilliant interval microscopic examination.
cresyl blue & crystal violet).  POSITIVE RESULT: 1. Sickle RBC (Hb S) but
does determine if heterozygous AS or
homozygous SS REPORTING: positive or
TESTS FOR SICKLING HEMOGLOBINS negative
SICKLE CELLS are distorted cells appearing as thin,  DEGREE of sickling depends on the
dense and elongated cells with both ends pointed. CONCENTRATION of the Hb S in the RBC
SICKLING is caused by the PRECIPITATION of an
 Readings are made at an HOURLY INTERVAL
ATYPICAL HEMOGLOBIN (commonly hemoglobin S)
for 2-3 HOURS
which DISTORTS the RED BLOOD CELLS into a SICKLE
 POSITIVE RESULT is DIAGNOSTIC but DOES
or CRESCENT SHAPE. HEMOGLOBIN S is fully soluble
NOT DISTINGUISH Hb S trait & Hb S anemia
when oxygenated but becomes insoluble when the
oxygen level is decreased. It first POLYMERIZES then
2. SODIUM METABISULFATE METHODS
forms into CRYSTALS which cause the red cell to
(DALAND & CASTLE)
become RIGID.
 REAGENT: 2% Sodium metabisulfide or
sodium bisulfate which are reducing agent
SCREENING TESTS:
which bind &remove O2 from the blood.
 PRINCIPLE: HbS forms an insoluble tactoid
 PRINCIPLE: a drop of blood is mixed w/ a
crystals when oxygen supply to the red cell is
drop of 2% sodium metabisulfite (a reducing
decreased. When Hb S is deoxygenized
agent) on a slide, & the mixture is sealed
outside the cell into the plasma it will cause
under a coverslip. The hemoglobin inside the
the solution to be TURBID. POSITIVE RESULT:
RBCs becomes deoxygenated causing
1. Sickle RBC 2. Turbidity
polymerization& the resultant sickle cell
 Degree of sickling or turbidity depends on the
formation
concentration of Hb S in the RBC
 PROCESS: add a drops of blood& the reagent
into the center of the slide.Cover the slide w/
HEMATOLOGY 2

coverslip&seal the sides. Observe  Abs are adsorbed to suspended metal sol
microscopically. particles giving the reagent raspberry-like
 POSITIVE RESULT: sickled RBC deoxygenated color
by reducing agent. NOTE:
 After electrophoresis, densitometry is
3. DITHIONITE SOLUBILITY TUBE TEST performed to determine the density of the
 REAGENT: sodium hydrosulfite (dithionite) bands created by the migrating proteins
removes O2 converted into concentration.
 SPECIMEN: whole blood w/ EDTA heparin or  In cellulose Acetate, at an alkaline pH, Hbs are
sodium citrate in order to prevent clotting negatively charge thus they migrate towards
 PRINCIPLE: Red cells are lysed by saponin the anode.Hb S together w/ D & G upon
allowing Hb to escape. Sodium dithionine migration appears as a band of protein thus
binds&removes oxygen from the test densitometry is performed to determine the
environment. Hb S polymerizes in the concentration of Hb S.
deoxygenated state&forms a precipitate in a  On Citrate Agar, at an acidic pH, Hb S migrates
high-molarity phosphate buffer solution. The towards the anode thus citrate agar is more
tactoid refract or deflect light& make the confirmatory
solution turbid.
 PROCESS: anticoagulated whole blood +
sodium dithionite. Saponin lyses the RBC EXPERIMENT MODULE:
releasing now Hb. Hb binds w/ sodium Some hemoglobins that aggregate and have reduced
dithionite which removes O2 from solution solubility are capable of polymerizing and crystallizing
causing Hb to crystalizes but polymerization within the red cell causing a distortion of cell shape
happens outside the RBC thus it will not (sickle shape). Hb S (Sickling Hb), when fully
cause sickling but turbidity of solution& is oxygenated is fully soluble. Polymerization and
determined by refraction&deflection of the formation into tactoid crystals occur only when
solution in the presence of light. Observe oxygen is decreased at tissue level.
specimen for turbidity by holding the tube  SPECIMEN NEEDED: Capillary blood
2.5 cm in front of a newsprint or a card
reader w/ thin black line A. SCRIVER AND WAUGH METHOD
 RULE: if the solution is observed in the a. Place a rubber band around the base of
solution then the solution is not turbid thus the middle finger and allow stayingin
negative result but if lines are no longer place for 5 minutes.
visible then it is positive for tactoid b. Make a finger puncture on the ball of the
crystals&sickling. finger and place a drop of capillary blood
 POSITIVE RESULT: turbidity on a slide.
 Turbidity indicates presence of sickling Hb c. Immediately cover with a coverslip and
regardless of any genotype seal edges with petroleum jelly.
 SCREENING METHOD OF CHOICE d. Incubate the preparation at room
temperature. Observe for red cell sickling
4. HEMOCARD Hb A & S at hourly intervals for 2, 3 hours, or after
 PRINCIPLE: Hb A& S contains monoclonal Ab 24 hours if desired.
(IgG), which specifically bind to the amino e. Microscopic examination (400x): If more
acids at or near the 6th position of the B- than 10% of the cells are sickled, the result
chain of the Hb S&A is positive.
 Hb S monoclonal Ab will react w/ the B-chain
of Hb S but not w/ the Hb A B. SICKLE CELL SLIDE TEST
 Hb A monoclonal Ab will react w/ the B-chain  In the absence of the HbS solubility test, the
of the Hb A but not w/ Hb S sickle cell slide test is useful in detecting
sickle cells in patients who have either sickle
cell disease or sickle cell trait.
HEMATOLOGY 2

a. Weigh 0.1g of sodium metabisulphite and OSMOTIC FRAGILITY TEST (OFT)


transfer to a test tube capable of holding  A test to identify the ability of red cells to
15 ml of water. absorb water without lysing.
b. Add 5 ml of distilled or deionized water,  It reflects the shape and size of erythrocyte
stopper, and mix until the chemical is (specifically the surface area-to-volume ratio)
fully dissolved. The chemical can only be  Not a confirmatory test for HS; it will only
used within the day it was suspended identify the presence of fragile cells like
(within 8 hrs) spherocytes. But it will not determine
c. Deliver one drop of patient’s capillary whether the spherocytosis is hereditary,
blood or well mix venous blood on a slide immune or other causes.
and add an equal volume of freshly made  For hereditary spherocytosis and other
reagent, mix and cover with a cover glass. anemia characterized by spherocytosis
Exclude any air bubbles. o Spherocytes are test using OFT
d. Place the slide in a plastic box or Petri because they are the most fragile cell
dish with damp piece of blotting paper or  able to detect the presence of
tissue at the bottom to prevent drying of spherocyte in the circulation
the preparation (moist chamber). Leave o RESULT IN HS: 0.60 - 0.65% saline
at room temperature. solution, hemolysis is already
e. After 10-20 minutes, examine the observed: spherocyte (fragile) easily
preparation microscopically for sickle lyse
cells. Focus the cells first with the 10x  PRINCIPLE: The cell is placed in a hypotonic
objective and examine for sickling using saline to cause osmosis (the shift of the fluid
the 40xobjective. Examine several fields. from the hypotonic saline into the red blood
Sickling usually takes place in one part of cell). If the red cell could still contain its
the preparation than the other. volume, the RBC would be able to absorb
f. Sickle cells usually appear crescent shape water without lysing and if not, it will easily
with pointed ends or holly leaf shape. hemolyse. POSITIVE RESULT: Hemolysis
Report as “sickle cell test positive” when  RESULTS:
crescent shape cells are seen, or ‘sickle o Incomplete hemolysis: 0.45%
cell test negative” when cells appear  Faint pink tinge supernatant with
rounded or oval shape. intact red cell button
o Complete hemolysis: 0.35%
C. TUBE SOLUBILITY TEST  Homogenous pink/red solution
 The principle of solubility method was based with the absence of red cell
on turbidity created when Hb S is incubated button
with sodium dithionate.  Fragility means increased OFT cells are easily
a. Add 20 uL of blood with 2 ml of the lyse (spherocytes)
reagent prepared in procedure B (2% w/v  INCREASED OFT:
sodium metabisulphite or sodium o HS, abnormal membrane, severe G-6-
dithionite) PDH deficiency, PK deficiency,
b. Mix and stand at room temperature for 5 Hemolytic anemia
minutes.  DECREASED OFT:
c. Examine the tubes using a white board o Resistant to red cell lysis even when
with black lines as the background in placed in hypotonic solution
ambient light settings.  With increased surface area-to
d. Interpret results as positive if the black volume ratio
lines are not visible  Hypochromic cells, target
cells/leptocytes, sickle cell
o Severe IDA (microcytic-hypochromic
cell)
HEMATOLOGY 2

o Thalassemia (microcytic-hypochromic o Determine the concentration of nacl


and target cell) solution where initial and complete
o Sickle cell anemia: If the sickling is still hemolysis occurred.
mild and is reversible, when the sickle o FORMULA: %NaCl = test tube
cell absorbs water it goes back to number x 0.02
being a normal cell. However, if its o PROCEDURE: Prepare different
membrane is totally damaged, the concentration of hypotonic saline
sickling becomes irreversible using the following procedure:
therefore becomes resistant. a. Set up 12 test tubes and label no.
 Affected by: 14 -25.
o Surface area-to-volume ratio b. Place in each tube 0.5% NaCl
 A normocyte with normal solution (The number of the tube
central pallor is resistant to corresponds to the drops of 0.5%
lysis because the space can NaCl)
bloat to extend the capacity, c. Using the same dropper, add
so the cell can absorb more distilled water into each tube
water without hemolyzing. until a total of 25 drops (NaCl &
But a cell without a central water) are in each tube.
pallor will immediately lyse d. Dispense to each tube one drop
upon absorption of water like of heparinized or defibrinated
spherocyte. blood.
 If the surface area-to-volume NOTE: Deliver at the same angle
ratio is still high, the cell is directly to saline solution.
resistant e. Mix tube and allow to stand at
 If the surface area-to-volume room temperature or centrifuge
ratio is decrease, the cell is for 1 minute at 2,500 rpm.
fragile  easily lyse f. Examine each tube for
 Cells with decreased surface initial/incomplete and complete
are-to-volume ratio have a hemolysis,
limited capacity to expand in
hypotonic solution and  DACIE METHOD  MORE ACCURATE
therefore lyse even at a less o Hypotonic saline (pH 7.4)
hypotonic concentration of  Buffered at pH 7.4 using PO4
saline than the normal in order to prevent
biconcave cells. interference of inconsistent
Ph
o Functional state of the cell  RECAP: OSMOSIS: movement
membrane of fluid in order to attain
 If the red cell membrane is homeostasis/equilibrium
defective, it is easily  Outside is hypotonic, inside is
hemolyse. isotonic  movement of
 Like in HS, there is membrane water from outside to inside
problem wherein it lack o HEMOLYSIS: @540 nm
integrity due to loss of o INCUBATED OFT: incubate the sample
protein. for at least 24 hours to detect mild
 SANFORD METHOD  MACROSCOPIC forms of HS
o REAGENT: 0.5% saline + add  It is done because if the
corresponding drops of DH2O observation is done
o SPECIMEN: Heparinized or immediate and the HS is mild
defibrinated blood there will be no hemolysis
seen  false negative result
HEMATOLOGY 2

 Mild forms of HS: on


prolonged incubation they
become fully spherocyte
therefore show hemolysis
 PURPOSE: detect mild forms
of HS.

You might also like