Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

OUTLINE • Shape (poikilocytosis-different shapes)


I. ANEMIA • In general:
o Microcytic hypochromic anemias are caused by disorders
A. Red Blood Cell Indices
of hemoglobin synthesis (so there is something to do with
B. Anemia of Blood Loss the production of hemoglobin), most commonly iron
C. Haemolytic Anemia deficiency anemia.
a. Hereditary Spherocytosis o Macrocytic anemia often stems from abnormalities that
b. Glucose-6-phosphate impair the maturation of erythroid precursors in the
Dehydrogenase Deficiency (G6PD) marrow. (Megaloblastic anemia)
c. Sickle Cell Disease o Normochromic normocytic anemias have diverse
etiologies (abnormal hemoglobin content but normal size)
d. Thalassemia Syndromes
• Red cells indices which are precisely measured can be used:
e. Paroxysmal Nocturnal o Mean Cell Volume (MCV) - the average volume of a red
Hemoglobinuria (PNH) cell, expressed in femtoliters (fL). NV: 80-100fL
f. Immunohemolytic Anemia ▪ <80 fL: Microcytic >100 f: Macrocytic
g. Hemolytic Anemia from Trauma to o Mean Cell Hgb (MCH) - average content (mass) of Hgb
Red Cells per RBC, expressed in pictograms (pg) NV: 23-35pg
D. Anemia of Diminished Erythropoiesis o Mean Cell Hgb concentration (MCHC) - average
concentration of Hgb in a given volume of packed RBC,
a. Megaloblastic
expressed in grams per deciliter (g/dL)
anemiaMegaloblastic anemia o Red cell distribution width - coefficient of variation of red
b. Pernicious anemia cell volume
c. Folic acid deficiency anemia ▪ The higher red ell distribution width, they are more
d. Iron deficiency anemia associated to aniso or poikilocytosis
e. Anemia of chronic inflammation
f. Aplastic anemia
g. Pure Red Cell anemia
h. Myelophthisic anemia

II. POLYCYTHEMIA
III. BLEEDING DISORDERS
A. Bleeding disorders caused by vessel wall
abnormalities
B. Thrombocytopenia
C. Bleeding disorders with defective
platelet function
D. Hemorrhagic diathesis related to
abnormalities in clotting factors
E. Disseminated Intravascular Coagulation
Table 1.1: Adult Reference ranges for RBC (10th Edition)
F. Complications of TransfusionBLEEDING
DISORDERS
IV. REFERENCES
V. TEST YOURSELF
VI. APPENDIX

I. ANEMIA
• For RBC and bleeding disorders, we assess your RBC of
course, your abnormality with the rbc. Your rbc is one of the
components of our blood. So what are the other components
of the blood? we have plasma, platelets, RBCs and WBCs.
• For bleeding disorders, we assess that with your platelets or
megakaryocytes.
• So your RBCs carry oxygen to the tissues, most common Table 1.2: Adult Reference ranges for RBC (9th Edition)
abnormality is anemia.
• Defined as the reduction of total circulating cell mass.
• It reduces the oxygen-carrying capacity of the blood leading to A. Red Blood Cell Indices
tissue hypoxia (lesser volume to RBCs leads to lesser volume • Used to define the size and hemoglobin content of the red blood
in the tissues leading to hypoxia) cell
• Usually, clinically diagnosed based on: • CONSISTS OF:
o hematocrit - ratio of packed RBC to total blood volume. o MCV- MEAN CORPUSCULAR VOLUME now called
o Hemoglobin concentration- protein that carries O2 mean cell volume
• Anemias can also be classified based on the alterations in red o MCH- MEAN CORPUSCULAR HEMOGLOBIN
cell morphology, which often point to particular cases. o MCHC- MEAN CORPUSCULAR HEMOGLOBIN
• Cell size (normo-, micro-, macrocytic) CONCENTRATION
• Degree of hemoglobinization (normo-, hypochromic)
Prepared by: CMED 2B Page 1 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

A. MEAN CELL VOLUME


The average volume of red cells and is calculated using the
following formula:

𝑯𝒄𝒕 𝒙 𝟏𝟎𝟑 𝒇𝑳
𝑴𝑪𝑽 =
𝑹𝑩𝑪/𝑳

MCV = volume of RBC in Femtoliters (fL) of blood


GIVEN:
Hematocrit = 45% or 0.45L
RBC = 5.0 x 1012/L
1 UL = 109 FL
1L = 1015 FL

Then
𝟎. 𝟒𝟓 𝒙 𝟏𝟎𝟏𝟓 𝒇𝑳/𝑳
𝑴𝑪𝑽 = Figure 3: Normocytic Anemia
𝟓. 𝟎 𝒙 𝟏𝟎𝟏𝟐 /𝑳
𝑴𝑪𝑽 = 𝟎. 𝟎𝟗 𝒙 𝟏𝟎𝟑 𝒇𝑳
B. MEAN CELL HEMOGLOBIN (MCH)
𝑴𝑪𝑽 = 𝟗𝟎 𝒇𝑳 Is the content (weight) of hemoglobin of the average red cell; it
is calculated from the hemoglobin concentration and the red cell
NORMAL Range: 80 to 100 fL
count:
𝑯𝑮𝑩 (𝑮/𝑳) 𝑷𝑮
MCV CLASSIFIES ANEMIA BASED ON RBC SIZE 𝑴𝑪𝑯 =
• <80 FL: MICROCYTIC ANEMIA 𝑹𝑩𝑪(/𝑳)
• 80-100: NORMOCYTIC ANEMIA
𝒘𝒆𝒊𝒈𝒉𝒕 𝒐𝒇 𝑯𝒈𝒃 𝒊𝒏 𝟏 𝑳 𝒐𝒇 𝒃𝒍𝒐𝒐𝒅
• >100: MACROCYTIC ANEMIA 𝑴𝑪𝑯 =
# 𝒐𝒇 𝑹𝒆𝒅 𝑪𝒆𝒍𝒍𝒔 𝒊𝒏 𝟏 𝑳 𝒐𝒇 𝒃𝒍𝒐𝒐𝒅

IF: 1G = 1012 PG
1L = 10 DL

Then:
𝑯𝑮𝑩 𝒙 𝟏𝟎 𝒙 𝟏𝟎𝟏𝟐 𝑷𝑮/𝑳
𝑴𝑪𝑯 =
𝑹𝑩𝑪/𝑳
IF: HGB = 15.0 g/dL
RBC = 5.0 x 1012/L

Then:
𝟏𝟓 𝒙 𝟏𝟎𝟏𝟑 𝑷𝑮/𝑳 𝟏𝟓 𝒙 𝟏𝟎 𝑷𝑮/𝑳
𝑴𝑪𝑯 = = = 𝟑𝟎𝑷𝑮
𝟓.𝟎𝒙 𝟏𝟎𝟏𝟐 /𝑳 𝟓.𝟎 𝒙 𝟏𝟎𝟏𝟐 /𝑳
Figure 1: Macrocytic Anemia
NORMAL RANGE FOR MCH = 27 to 31 PG

C. MEAN CELL HEMOGLOBIN CONCENTRATION (MCHC)


Is the average concentration of Hemoglobin in a given volume
of packed red cells:
𝐻𝐺𝐵 𝑥 100% 𝐻𝐺𝐵 𝑔/𝑑𝐿
𝑀𝐶𝐻𝐶 = 𝑜𝑟
𝐻𝐶𝑇 𝐻𝐶𝑇
𝐻𝐺𝐵 𝑖𝑛 𝑔/𝑑𝐿
𝑀𝐶𝐻𝐶 = 𝑥 100 (𝑡𝑜 𝑐𝑜𝑛𝑣𝑒𝑟𝑡 𝑡𝑜 %)
𝐻𝐶𝑇(𝑣𝑜𝑙𝑢𝑚𝑒 𝑜𝑓 𝑅𝐵𝐶 𝑖𝑛 𝑔/𝑑𝐿

IF: HGB = 15.0 g/dL


HCT = 45% (0.45)

THEN:
15.0 𝑔/𝑑𝐿 15.0 𝑔/𝑑𝐿
𝑀𝐶𝐻𝐶 = 𝑥 100 = 𝟑𝟑. 𝟑% 𝒐𝒓 = 𝟑𝟑. 𝟑 𝒈/𝒅𝑳
0.45 𝑔/𝑑𝐿 0.45

Therefore, the Formula


Figure 2: Microcytic Anemia 𝐻𝐺𝐵 𝑥 100% 𝐻𝐺𝐵 𝑔/𝑑𝐿
(associated with iron deficiency anemia) 𝑀𝐶𝐻𝐶 = 𝑜𝑟
𝐻𝐶𝑇 𝐻𝐶𝑇

NORMAL RANGE for the MCHC: 31% to 36 % = 31 to 36 g/dL


• <31% = Hypochromia
• 31%-36% = Normochromia
• >36% = Hyperchromia associated to megaloblastic anemia

Prepared by: CMED 2B Page 2 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

Reticulocytes are larger than normal red cells and


have blue-red polychromatophilic cytoplasm due to
the presence of RNA, a feature that allows them to be
identified in the clinical laboratory. Early recovery
from blood loss also is often accompanied by
thrombocytosis, which results from an increase in
platelet production.

2. Chronic Blood Loss


• Induces anemia only when the rate of loss exceeds the
regenerative capacity of the marrow.
• It can also cause anemia when iron reserves are depleted
and iron-deficiency anemia (IDA) appears.

C. Hemolytic Anemia
• Hemolysis- Breakdown of RBCs
• Common features of hemolytic anemias:
• Shortened red cell life span below 120 days.
• Elevated erythropoietin levels and compensatory increase in
Figure 4: Hypochromia erythropoiesis.
• Accumulation of Hgb degradation products that are created as
B. Anemia of Blood Loss a part of red cell hemolysis
• Persistent hemolysis leads to hyperplasia of phagocytes
• Regardless of cause, severe anemia leads to certain clinical manifested by varying degrees of splenomegaly.
findings: • The physiologic destruction of senescent red cells takes place
o Patients appear pale (Pallor) within macrophages.
o Weakness, Malaise, and Easy Fatigability it’s because of • Abundant in the spleen, liver, and bone marrow.
the decrease oxygen delivery in the body. • This process appears to be triggered by age-dependent
o Dyspnea on mild exertion due to decreased 02
changes in red cell surface proteins, which lead to their
o Hypoxia can cause fatty change in liver, myocardium,
recognition and removal by phagocytes.
kidney
• In the great majority of hemolytic anemias, the premature
• Fatty changes in the myocardium can cause cardiac failure.
destruction of red cells also occurs within phagocytes, an event
Hypoxia in the myocardium often manifests as angina pectoris
that is referred to as extravascular hemolysis. If persistent,
• Acute blood loss and shock, oliguria or anuria can develop as extravascular hemolysis leads to a hyperplasia of phagocytes
a result of renal hypoperfusion. (no oxygen in the kidneys) manifested by varying degrees of splenomegaly.
• CNS hypoxia can cause headache, dimness of vision,
faintness. 1. Extravascular Hemolysis
• Caused by alterations that render the RBC less
1. Acute Blood Loss deformable in passing the splenic sinusoids.
• Effects are mainly due to loss of intravascular volume, • This causes red cell sequestration and phagocytosis.
which if massive loss of blood, can lead to cardiovascular
• Principal features are:
collapse, shock, and death.
o Anemia (pallor)
• Reduction of 02 triggers increased secretion of o Splenomegaly (increase in size of the spleen)
erythropoietin from the kidneys, stimulating proliferation of o Jaundice (yellowing of the skin)
committed erythroid progenitors.
• Some hemoglobin inevitably escapes from phagocytes,
o It takes 5 days for them to completely mature and which leads to variable decreases in plasma haptoglobin,
appear as reticulocytes in the peripheral blood.
an α2-globulin that binds free hemoglobin and prevents its
o If there is increased reticulocytes in the blood, it
excretion in the urine.
signals that there is increase loss of RBCs..The
lifespan of RBCs is 120 days, so if there is increased
2. Intravascular Hemolysis
reticulocytes in the blood, there is increased turnover
• May be caused by mechanical injury (trauma to cardiac
of RBC to shorten their lifespan.
valves), complement fixation, intracellular parasites
o The iron in hemoglobin is recaptured if red cells
(malaria), or exogenous toxic factors (clostridial sepsis), it
extravasate into tissues, whereas bleeding into the
is also associated with incompatibility in blood transfusion.
gut or out of the body leads to iron loss and possible
iron deficiency, which can hamper the restoration of • Manifests as:
normal red cell counts. o Anemia
o Significant bleeding results in predictable o Hemoglobinemia (blood plasma will be reddish or
changes in the blood involving not only red cells, pinkish)
o Hemoglobinuria
but also white cells and platelets. If the bleeding is
o Hemosiderinuria
sufficiently massive to cause a decrease in blood
o Jaundice
pressure, the compensatory release of adrenergic
hormones mobilizes granulocytes from the • Morphology:
intravascular marginal pool and results in o Increased number of erythroid precursors
leukocytosis. Initially, red cells appear normal in size (normoblasts) the younger cells of erythropoiesis.
and color (normocytic, normochromic). However, as o Compensatory increase in erythropoiesis leading to
marrow production increases, there is a striking prominent reticulocytosis in the peripheral blood.
increase in the reticulocyte count (reticulocytosis), (Increased number of retics)
which reaches 10% to 15% after 7 days. o Accumulation of hemosiderin in the spleen, marrow,
liver.
Prepared by: CMED 2B Page 3 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

o Accumulation of this iron pigment is called • The pathogenic mutations most commonly affect ankyrin, band
hemosiderosis 3, spectrin, or band 4.2, the proteins involved in one of the two
o Severe anemia can lead to extramedullary tethering interactions.
hematopoiesis spleen, liver, or lymph nodes. Usually, • Most mutations cause frameshifts or introduce premature stop
hematopoiesis takes place in the bone marrow but codons, such that the mutated allele fails to produce any
when there is extravascular hemolysis, there is protein.
hematopoiesis in the spleen, liver or lymph nodes. • The resulting deficiency of the affected protein reduces the
o Pigment gallstones can also form due to elevated assembly of the skeleton as a whole, destabilizing the
biliary excretion of bilirubin in chronic hemolysis. overlying plasma membrane.
(black gallstones • Because of the spheroidal shape and reduced deformability,
o Iron released from hemoglobin can accumulate within the Shapeless spherocytes are trapped in the splenic cord.
tubular cells, giving rise to renal hemosiderosis. • Splenic environment exacerbates loss of membranes together
o Heme groups derived from hemoglobin-haptoglobin with K+ and H20.
complexes are metabolized to bilirubin within • Prolonged splenic exposure (erythrostasis).
mononuclear phagocytes, leading to jaundice. • Depletion of glucose and diminished red cell pH. (acidic)
• After splenectomy, anemia is corrected but spherocytes still
a. Hereditary Spherocytosis persists.
• HS is an inherited disorder caused by intrinsic defects in the • Spherocytosis apparent on smears as small, dark-staining
red cell membrane skeleton (spectrin and ankyrin), rendering it (hyperchromie) red cells lacking the central zone of pallor.
spheroid, less deformable, and prone to splenic sequestration. • Distinctive but not pathognomonic, as they are also seen in
• Highest prevalence in Northern Europe (1 in 5,000). disorders with membrane loss such as autoimmune hemolytic
• Autosomal Dominant disorder in 75% of cases. anemia
• Remaining has more severe disease caused by two different • Other features common to hemolytic anemias
defects, a state known as compound heterozygosity. • Hemosiderosis, Reticulocytosis, Marrow erythroid hyperplasia,
mild jaundice.
• Cholelithiasis in 40-50% of adults. (pigmented gallstones)
• Moderate splenomegaly (500-1000g) due to congestion.
• The diagnosis is based on family history, hematologic findings,
and laboratory evidence.

• Characteristic features:
o Anemia
o Splenomegaly
o Jaundice
Clinical signs are usually the same with other anemias so how
are we going to differentiate them? we differentiate them using
CBC, we assess it and further seen in your peripheral blood
smear. So we diagnose anemias manually using your
microscope by their differences in RBC morphology.
• Small population present with marked jaundice at birth
requiring exchange transfusion.
• 20-30% is so mild to be virtually asymptomatic.
• Generally, Stable clinical course is sometimes punctuated by
aplastic crises, triggered by acute parvovirus infection.
o Parvovirus B19 (Erythema infectious) infects and kills red
cell progenitors, causing red cell production to cease until
immune response commences (1-2 weeks).
• Due to decreased life span, anemia can worsen, transfusion
may be necessary until immune response clears infection
• Transfusions may be necessary to support the patient
during the acute phase of the infection.
• Hemolytic Crises produced by intercurrent events leading to
increased splenic destructions (e.g. Infectious mono) but less
significant than aplastic crises.

Figure 5: Pathophysiology of Hereditary Spherocytosis

• HS is caused by diverse mutations that lead to an


insufficiency of membrane skeletal components.
• As a result of these alterations, the life span of affected red cells
is decreased on average to 10 to 20 days from the normal 120 Figure 6: Hereditary Spherocytosis
days.

Prepared by: CMED 2B Page 4 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

b. Glucose-6-phosphate Dehydrogenase o Since hemolysis is episodic, features to chronic hemolysis


are absent (e.g. splenomegaly, cholelithiasis).
Deficiency (G6PD)
• Abnormalities in the Hexose monophosphate shunt or
glutathione metabolism resulting from deficient or impaired
enzyme function reduce the ability of RBC to protect
themselves from oxidative injury and lead to hemolysis.
• The most important of these enzyme derangements is
hereditary deficiency of glucose-6-phosphate dehydrogenase
(G6PD) activity.
• G6PD reduces nicotinamide adenine dinucleotide phosphate
(NADP) to NADPH while oxidizing glucose-6-phosphate.
NADPH then provides reducing equivalents needed for
conversion of oxidized glutathione to reduced glutathione,
which protects against oxidant injury by participating as a
cofactor in reactions that neutralize compounds such as H2O2.
• G6PD deficiency is a recessive, X-linked disorder, posing
higher risk for males. Several variants, but most are harmless
• Most clinically significant are:
• Designated G6PD-: present in 10% of Americans.
• G6PD Mediterranean: common in Middle East
• Because mature red cells do not synthesize new proteins, as
red cells age G6PD– and G6PD Mediterranean enzyme
activities quickly fall to levels that are inadequate to protect
against oxidant stress.
• Older red cells are much more prone to hemolysis than younger
ones.
• Believed to stem from protective effects against Plasmodium
falciparum malaria
• Episodic hemolysis is characteristic of G6PD deficiency is
caused by exposures that generate oxidative stress.
• Most common triggers:
• Infections
o Radicals produced by macrophages
o Most common.
o Viral hepatitis, pneumonia, typhoid fever so if there is
infection there will be hemolysis.
• Drugs:
o Anti-malarial drugs (primaquine, chloroquine)
o Sulfonamides, Nitrofurantoins, etc.
• Fava beans, most frequently cited oxidant producer
o Uncommonly, G6PD deficiency presents with neonatal
jaundice or a chronic low-grade hemolytic anemia in the
absence of infection or known environmental trigger.
• Oxidants cause both intra- and extravascular hemolysis
• This is not a dangerous deficiency because in can be prevented
it’s just that don’t eat and take drugs that can cause trigger for
hemolysis and this can be evaluated in your newborn screening
so this can be prevented. This can be readily tested upon
delivery of the child.
• Morphologically what we usually see in the blood smear is the
presence of Heinz bodies, Heinz bodies can only be seen using
a supravital stain likewise your reticulocytes we use supravital
stain. Heinz bodies are formed due to exposure of G6PD
deficient cells to high levels of oxidants, causing cross-linking
Figure 7: G6PD Deficiency
of reactive sulfhydryl groups on globin chains that become
denatured c. Sickle Cell Disease
• As the red cells pass the spleen, macrophages pluck out the • A common hereditary hemoglobinopathy caused by a point
Heinz bodies causing membrane damage with partially mutation in the ß-globin that promotes the polymerization of
devoured shape called Schistocytes (Bite cells). deoxygenated hemoglobin, leading to red cell distortion,
• Clinical Features hemolytic anemia, microvascular obstruction, and tissue
o Acute intravascular hemolysis, marked by anemia, damage.
hemoglobinemia, and hemoglobinuria 2-3days following • Sickle cell disease is caused by a missense mutation in the β-
exposure to oxidants. globin gene that leads to the replacement of a charged
o Episode is self-limited: □ Only older cells are at risk for glutamate residue with a hydrophobic valine residue.
lysis, hemolysis stops when only younger cells remain in • The abnormal physiochemical properties of the resulting sickle
the blood. hemoglobin (HbS) are responsible for the disease.
o Recovery Phase is heralded by reticulocytosis. • Occurs primarily in individuals of African descent.

Prepared by: CMED 2B Page 5 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• The high prevalence of sickle cell trait in certain African • Intracellular dehydration increases MCHC, facilitates
populations stems from its protective effects against falciparum sickling. So if there is dehydration, there is increased
malaria. hemoglobin concentration so they are more prone to
• Sickle Cell trait - about 8-10% of African Americans (2 million) hypoxia.
are heterozygous for HbS, which presents as asymptomatic • Conditions that decrease MCHC reduce the disease
• The point mutation in the 6th codon of ß-globin leads to the severity
replacement of a glutamate residue with a valine residue.
• The abnormal physicochemical properties of the resulting 3. Intracellular pH
sickle cell hemoglobin (HbS) are responsible for the disease. • Decrease in pH reduces 02 affinity, thereby increasing the
• High frequency stems from protection afforded by HbS against fraction of deoxygenated HbS and may augment sickling.
falciparum malaria, 6 times increase in areas in which malaria More acidity leads to more sickling
is endemic.
• MORPHOLOGY:
o Peripheral blood demonstrates irreversibly sickled cells,
reticulocytosis, and target cells, resulted from dehydration.
o Howell-Joly Bodies (small nuclear remnants) are also
present due to asplenia.
o There is erythroid hyperplasiam leading to bone marrow
expansion resulting to bone resorption and secondary new
bone formation, resulting to prominent cheek bones and
changes in the skull that resembles a crewcut.
o Extramedullary hematopoiesis
o The increased breakdown of hemoglobin may cause
hyperbilirubinemia and formation of pigment gallstones.

• CLINICAL FEATURES:
o Moderately severe hemolytic anemia (Hct: 18-30%), with
reticulocytosis, hyperbilirubinemia, and irreversibly sickled
cells.
o Vaso-occlusive crises, also called pain crises, are
episodes of hypoxic injury and infarction that cause severe
pain in the affected region
✓ Most commonly involved are bones, lungs, liver,
brain, spleen, and penis.
✓ Frequently manifests as hand-foot syndrome or
dactylitis.
✓ Acute chest syndrome involves lungs, presents as
fever, cough, chest pain, and inflammatory infiltrates.
✓ Stroke

• Aplastic Crises
o Stem from infection of red cell progenitors by
Parvovirus B19, causing transient cessation of
erythropoiesis, leading to anemia.
• Chronic Tissue Hypoxia
Figure 8: Pathophysiology of Sickle Cell Disease o Generalized impairment of growth and development
as well as organ damage
• G6PD deficiency and thalassemia also protect against
malaria by increasing the clearance and decreasing the • Increased susceptibility to encapsulated organisms due to
adherence of infected red cells, possibly by raising levels altered splenic function.
of oxidant stress and causing membrane damage in the • Pneumococcus pneumonia, and Haemophilus influenzae
parasite-bearing cells that leads to their rapid removal septicemia and meningitis are common (encapsulated
from the bloodstream. organisms: S. pneumonia, K. pneumoniae, B. anthracis)
• Mixing a blood sample with an oxygen consuming agent
(metabisulfite) induces sickling of red cells.
• About 90% of patients survive to age 20, and 50% survive
Variable Affecting the degree and Rate of Sickling: beyond the 5th decade.
• Priapism affects up to 45% of males after puberty and may
1. Interaction of HbS with other types of haemoglobin lead to hypoxic damage and erectile dysfunction.
• In heterozygotes with sickle cell trait, 40% are HbS and the • Other disorders related to vascular obstruction, particularly
rest is HbA. The HbA interferes with the polymerization of stroke and retinopathy leading to loss of visual acuity and even
HbS thus cells do not sickle unless in states of hypoxia. blindness, can take a devastating toll.
• HbF inhibits polymerization more than HbA. HbA is your • Factors proposed to contribute to stroke include the adhesion
normal hemoglobin composed of 2 alpha and 2 beta globin of sickle red cells
chains.
• HbC, lysine is substituted for glutamate, compounds with • Treatment:
HbS forming HbSC red cells. o Mainstay treatment is an inhibitor of DNA synthesis
(Hydroxyurea) which has several benefits.
2. Mean Cell Hemoglobin Concentration (MCHC) o Increase in red cell HbF levels.
o Anti-inflammatory effect
Prepared by: CMED 2B Page 6 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

o In severe β-thalassemia, ineffective erythropoiesis creates


several additional problems.
o Erythropoietic drive in the setting of severe
uncompensated anemia leads to massive erythroid
hyperplasia in the marrow and extensive extramedullary
hematopoiesis.
o The expanding mass of red cell precursors erodes the
bony cortex, impairs bone growth, and produces skeletal
abnormalities (described later).
o Extramedullary hematopoiesis involves the liver, spleen,
and lymph nodes, and in extreme cases produces
extraosseous masses in the thorax, abdomen, and pelvis.
o The metabolically active erythroid progenitors steal
nutrients from other tissues that are already oxygen-
starved, causing severe cachexia in untreated patients.
Figure 9: Sickle cell o Erythroid precursors secrete a hormone called
(they lost already their membrane, the ankyrin and spectrin) erythroferrone that inhibits production of hepcidin, a key
negative regulator of iron uptake in the gut.
o In thalessemia, the marked expansion of erythroid
d. Thalassemia Syndromes precursors leads to increased absorption of iron from the
o Heterogeneous group of disorders caused by inherited gut, and this together with repeated blood transfusions
mutations that decrease the synthesis of either the a-globin or inevitably lead to severe iron accumulation (secondary
the ß-globin chains that compose the adult Hgb: HbA (a2ß2). hemochromatosis) unless preventive steps are taken.
o 2 a-chains encoded by 2 a-globin genes on chromosome 16. Injury to parenchymal organs, particularly the heart and
o 2 ß-chains encoded by 1 ß-globin gene on chromosome 11. liver.
o So when we say a-thalassemia, there is abnormality or missing
in the alpha chain and when we say ß-thalassemia the o Clinical Syndromes
abnormality or missing is the beta chain. o The relationship depends on the severity of anemia
which in turn depends on the genetic defect (ß + or ß)
o ß-Thalassemia and the gene dosage (homozygous or heterozygous).
o Mutations that diminish the synthesis of ß-globin chains Of course, it is more severe when it is hmomozygous.
o Pathogenesis:
o Two categories of mutations: o ß-Thalassemia Major (ß+ ß+ or ßOßO or ß+ßO)
▪ ßo mutations- absent ß-globin synthesis o Two ß-thalassemia alleles (Homozygous) have severe
▪ ß+ mutations - reduced ß-globin synthesis transfusion-dependent anemia.
o 100 different mutations, mostly point mutations: o Most common in Mediterranean, parts of Africa, and south
o Splicing mutations East Asia. So in the Philippines, we see patients with
o Most common cause of ß+ mutations thalassemia.
o Some of these mutations destroy normal RNA splice o Manifests 9 months after birth as HbF switched to HbA.
junctions and completely prevent the production of o PBS: Anisiopoikilocytosis, Reticulocytosis, Schistocytes,
normal β-globin mRNA, resulting in β0 -thalassemia. Target Cells.
o Promoter Region Mutations o It is also included in the newborn screening so as early as
o Associated with ß+ mutations. baby upon delivery, you could determine if the patient has
o Chain Terminator Mutations thalassemia unlike before we have to wait until night
o Most common cause of ßO mutations. months for the manifestations. Prevention is earlier too
o They consist of either nonsense mutations that now.
introduce a premature stop codon or small insertions o If you notice, their appearances in the blood is the same
or deletions that shift the mRNA reading frames. so we diagnose your RBC abnormalities/defects using
CBC when you see there is flagging with the use of the
o Impaired β-globin synthesis results in anemia by two RBC indices and RDW we make smears and view that in
mechanisms. the microscope but they have the same RBC variations
o The deficit in HbA synthesis produces and inclusions so to differentiate them, for thalassemia for
“underhemoglobinized” hypochromic, microcytic red confirmatory we have to undergo electrophoresis which is
cells with subnormal oxygen transport capacity. a higher and mor sophisticated machine for thalassemia.
o Even more important is the diminished survival of red
cells and their precursors, which results from the o ß-Thalassemia minor/trait (ß+ß or ßOß)
imbalance in α- and β-globin synthesis. o More common than major, mild, asymptomatic anemia.
o Unpaired α chains precipitate within red cell precursors, o Abnormal PBS, mild erythroid hyperplasia
forming insoluble inclusions. These inclusions cause a o ß-Thalassemia intermedia - milder forms of ß+ß+. ß+ßO.
variety of untoward effects, but membrane damage is the o The peripheral blood smear typically shows hypochromia,
proximal cause of most red cell pathology. microcytosis, basophilic stippling, and target cells.
o Many red cell precursors succumb to membrane damage o Mild erythroid hyperplasia is seen in the bone marrow.
and undergo apoptosis. o Hemoglobin electrophoresis usually reveals an increase in
o In severe β-thalassemia, it is estimated that 70% to 85% HbA2 (α2δ2) to 4% to 8% of the total hemoglobin (normal,
of red cell precursors suffer this fate, which leads to 2.5% ± 0.3%), reflecting an elevated ratio of δ-chain to β-
ineffective erythropoiesis. chain synthesis.
o Those red cells that are released from the marrow also o HbF levels are generally normal or occasionally slightly
contain inclusions and have membrane damage, leaving increased.
theme prone to splenic sequestration and extravascular o Recognition of β-thalassemia trait is important for two
hemolysis. reasons:
1. it may be mistaken for iron deficiency
Prepared by: CMED 2B Page 7 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

2. it has implications for genetic counseling.


a-Thalassemia
o Iron deficiency (the most common cause of microcytic • Inherited deletions that result in reduced or absent synthesis of
anemia) can usually be excluded by measurement of a-globin chains.
serum iron, total iron-binding capacity, and serum ferritin. • Normally there are 4 a-globin genes, the severity of a
o The increase in HbA2 is diagnostically useful, particularly thalassemia depends on the number of a-globin chains
in individuals (such as women of childbearing age) who mutated, it depends on the number of a-globin chains that have
are at high risk of iron deficiency. undergone mutations.
• The anemia stems from lack of adequate Hgb and presence of
• Blood smear shows severe red cell abnormalities: excess unpaired globin chains.
o Anisocytosis - change in size • In newborns, excess unpaired y-globin forming y4 tetramers
o Poikilocytosis - change in shape. known as Hemoglobin Barts.
• It also shows microcytosis and hypochromia. • Older children and adults, excess ß-globins form ß4 tetramers
• Target cells, basophilic stippling, and fragmented RBC known as HbH.
• Diagnosis of ß-Thalassemia Trait/Minor • If HbH that is your Beta-4 tetramers if hemoglobin Barts, that is
• Superficially resembles the hypochromic microcytic anemia of your y-4 tetramers so remember your normal lobin chain is
Iron Deficiency. alpha and beta so ther is a replacement of your globin chains.
• Implications for genetic counseling • Silent Carrier State
o Deletion of a single a-globin gene, which causes a barely
detectable reduction in a-globin synthesis.
o Completely asymptomatic with slight microcytosis.
o They are just carrier

Hemoglobin H Disease
• Caused by deletion of 3 a-globin genes.
• Most common in Asians.
• With only one normal a-globin gene, synthesis is markedly
reduced, and tetramers of ß-globins form.
• Moderately severe anemia, resembling ß- thalassemia
intermedia. It is severe but does not require blood transfusions
unlike your beta-thalassemia major.

Figure 10: Jigsaw Puzzle RBC

Table 2: Clinical and Genetic Classification of Thalassemias


*See Larger Image in Appendix

Hydrops Fetalis
• Most severe form of a-thalassemia, caused by deletion of all
four a-globin genes.
• In the fetus, y globins form tetramers (Hgb Barts) that also have
high affinity to oxygen causing tissue hypoxia.
• Fetus shows severe pallor, generalized edema, and massive
hepatosplenomegaly.
• Survival in early development is due to the expression of ζ
chains, an embryonic globin that pairs with γ chains to form a
functional ζ2γ2 Hb tetramer.
• Signs of fetal distress usually become evident by the third
trimester of pregnancy.
• With intrauterine transfusion many affected infants are now
saved.
• The fetus shows severe pallor, generalized edema, and
massive hepatosplenomegaly similar to that seen in hemolytic
disease of the newborn

e. Paroxysmal Nocturnal Hemoglobinuria (PNH)


• Results from acquired mutations in the phosphatidylinositol
glycan complementation group A gene (PIGA), an enzyme
essential for the synthesis of certain membrane-associated
complement regulatory protein.
• Thrombosis is the leading cause of disease-related death in
Figure 11: Codocytes or Target Cells
PNH.
(most commonly associated to thalassemia)
Prepared by: CMED 2B Page 8 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• 40% suffer from venous thrombosis, often involving hepatic • Cold Agglutinin Type
portal or cerebral veins. o Caused by IgM antibodies that bind to red cells avidly at
• 50% develop acute myelogenous leukemia or myelodysplastic low temperatures (OoC to 4oC).
disorder. o Less common than warm, accounts for 15-30% of cases.
• Diagnosis: Flow Cytometry (Detects GPI-deficient RBC). o Occurs after certain infections such as Mycoplasma
Sophisticated machine that diagnose PNH pneumonia, EBV, CMV, Influenza virus, HIV.
• Treatments: o Chronic cold agglutinin immunohemolytic anemia occurs
o Bone Marrow Transplantation, the only hope for cure in association with certain B-cell neoplasms.
o Infusion of monoclonal antibody inhibitor of C5a that o Occurs in exposed fingers, toes, or ears where vascular
reduces hemolysis, but can predispose to fatal beds’ temperature may fall below 30oC.
meningococcemia due to MAC deficiency. o Manifests as pallor, cyanosis, and Raynaud phenomenon
o Immunosuppressive drugs for those with marrow aplasia

Cold Hemolysin Type


f. Immunohemolytic Anemia • Autoantibodies that is responsible for an Paroxysmal Cold
Hemoglobinuria, a rare disorder that causes substantial,
sometimes fatal, intravascular hemolysis and hemoglobinuria.
• Autoantibodies are IgGs that bind to the P blood group antigen
in cool peripheral areas
• Most seen in children following viral infections, which recover
within 1 month.
• Treatment:
o Warm - removal of initiating factors such as the drugs,
immunosuppressive drugs and splenectomy.
o Chronic cold - more difficult to treat.

g. Hemolytic Anemia from Trauma to Red Cells


• Most significant is seen in individuals with cardiac valve
Table 3: Classification of Immunohemolytic Anemias prostheses and microangiopathic disorders.
• Artificial mechanical valves are more frequently implicated than
• Warm Antibody Type are bioprosthetic porcine valves (from a pig).
o Most common form. • Hemolysis stems from shear forces produced by turbulent
o This form constitutes approximately 80% of cases of blood flow and pressure gradients across damaged vessels
immunohemolytic anemia. It is caused by antibodies that • Microangiopathic hemolytic anemia is commonly seen in
bind stably to red cells at 37°C disseminated intravascular coagulation (DIC), but it can also
o 50% are idiopathic (Primary), others are related to occur in:
predisposing conditions (refer to table 14-4), or exposure o Thrombotic thrombocytopenic purpura (TTP)
to a drug. o Hemolytic uremic syndrome (HUS)
o Most causative are of the IgG class, less commonly IgA. o Malignant HTN, SLE, and disseminated cancer
Lysis is mostly extravascular. • These changes produce shear stress that leads to appearance
o Splenomegaly, many cases, antibodies are directed of red cell fragments (schistocytes), burr cells, helmet cells, and
against the Rh blood antigens. triangle cells.
o As with other autoimmune disorders, the cause of primary
immunohemolytic anemia is unknown.
o In cases that are idiopathic, the antibodies are directed
against red cell surface proteins, often components of the
Rh blood group complex.
o In drug-induced cases, two mechanisms have been
described:
▪ Antigenic drugs. Hemolysis usually follows large,
intravenous doses of the offending drug and occurs 1 to
2 weeks after therapy is initiated. These drugs,
exemplified by penicillin and cephalosporins, bind to the
red cell membrane and create a new antigenic
determinant that is recognized by antibodies. The Figure 11: Microangiopathic hemolytic anemia. A peripheral blood
responsible antibodies sometimes fix complement and smear from a person with hemolytic-uremic syndrome shows
cause intravascular hemolysis, but more often they act as several fragmented red cells
opsonins that promote extravascular hemolysis within
phagocytes. D. Anemias of Diminished Erythropoeisis
▪ Tolerance-breaking drugs. These drugs, of which the • Most common and important anemia associated with red cell
antihypertensive agent α-methyldopa is the prototype, underproduction is those caused by nutritional deficiencies.
break tolerance in some unknown manner that leads to • Followed by those that arise form Renal Failure and Chronic
the production of antibodies against red cell antigens, Inflammation.
particularly the Rh blood group antigens. About 10% of • Less common disorders that lead to generalized bone marrow
patients taking α-methyldopa develop autoantibodies, as failure:
assessed by the direct Coombs test, and roughly 1% o Aplastic Anemia
develop clinically significant hemolysis. o Primary hematopoietic neoplasm
o Infiltrative Disorder
Prepared by: CMED 2B Page 9 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

a. Megaloblastic Anemia • Neutrophils are also larger than normal and show nuclear
hypersegmentation, having five or more nuclear lobules
instead of the normal three to four.
• The marrow is usually markedly hypercellular as a result
of increased numbers of hematopoietic precursors.
• Megaloblastic changes are detected at all stages of
erythroid development.
• The most primitive cells (promegaloblasts) are large, with
a deeply basophilic cytoplasm, prominent nucleoli, and a
distinctive, fine nuclear chromatin pattern.
• As these cells differentiate and begin to accumulate
hemoglobin, the nucleus retains its finely distributed
chromatin instead of developing the clumped pyknotic
chromatin typical of normoblasts.
• Although nuclear maturation is delayed, cytoplasmic
maturation and hemoglobin accumulation proceed at a
normal pace, leading to nuclear-to-cytoplasmic
asynchrony.
• Because DNA synthesis is impaired in all proliferating
cells, granulocytic precursors also display dysmaturation
in the form of giant metamyelocytes and band forms.
• Megakaryocytes also may be abnormally large and have
bizarre, multilobate nuclei.

b. PERNICIOUS ANEMIA
• A specific form of megaloblastic anemia caused by an
autoimmune gastritis that impairs the production of
Intrinsic factor, which is required for B12 uptake from the
gut.
• Vitamin B12 requires IF secreted by parietal cells.
• Three types of autoantibodies responsible for the
autoimmune attack in the gastric mucosa:
o Type I - 75% blocks the binding of B12 to IF.
o Type II - Prevents binding of IF-B12 complex to lleal
receptor
o Type III - 85-90% recognize the a and B subunit of
the gastric proton pumps.

Table 4: Causes of Megaloblastic Anemia MORPHOLOGY


• Bone marrow and blood findings are same for all
• Vitamin B12 and B9 are coenzymes required for the megaloblastic anemias.
synthesis of thymidine, one of 4 bases of DNA. • Fundic Gland Atrophy, affecting both chief and parietal
• Deficiency or impairment in metabolism results in cells, there is intestinalization (metaplasia) or replacement
defective nuclear maturation due to deranged or of the glandular epithelium by mucus-secreting goblet cells
inadequate DNA synthesis, with an attendant delay or that resemble those lining the large intestine, a form of
block in cell division. metaplasia referred to as intestinalization.
• With time, the tongue may take on a shiny, glazed, “beefy”
TWO PRINCIPAL TYPES: appearance (atrophic glossitis).
a. Pernicious - major form of Vit. B12 deficiency anemia • Because the gastric atrophy and metaplastic changes are
b. Folate Acid deficiency anemia due to autoimmunity, they persist following parenteral
administration of vitamin B12, whereas the “megaloblastic”
MORPHOLOGY changes in the marrow and gut are readily reversible.
• Presence of red cells that are macrocytic and oval • Central nervous system lesions are found in about three-
(macroovalocytes) with anisopoikilocytosis is highly fourths of all cases of florid pernicious anemia but can also
characteristic. be seen in the absence of overt hematologic findings.
• Neutrophils are also larger and show nuclear hyper • The principal alterations involve the cord, where there is
segmentation, having >5 segments. (normal: 3-5 demyelination of the dorsal and lateral spinal tracts,
segmentations) sometimes followed by loss of axons. These changes may
• Marrow is markedly hypercellular with megaloblastic red give rise to spastic paraparesis, sensory ataxia, and
cell precursors, granulocytic precursors, and severe paresthesias in the lower limbs.
megakaryocytes. • Less frequently, degenerative changes occur in the
• Ineffective erythropoiesis due to apoptosis of precursor ganglia of the posterior roots and in peripheral nerves.
cells.
• There is marked variation in red cell size (anisocytosis) CLINICAL FEATURES
and shape (poikilocytosis). • Pernicious anemia is insidious in onset, and the anemia is
• The reticulocyte count is low. often quite severe by the time it comes to medical
• Nucleated red cell progenitors occasionally appear in the attention.
circulating blood when anemia is severe.

Prepared by: CMED 2B Page 10 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• The course is progressive unless halted by therapy. The 3. derangements associated with hyperactive
diagnosis is based on: hematopoiesis (e.g., chronic hemolytic anemia)
1. a moderate to severe megaloblastic anemia 4. disseminated cancer.
2. leukopenia with hypersegmented granulocytes • In all of these circumstances, the demands of increased
3. low serum vitamin B12 DNA synthesis render normal intake inadequate.
4. elevated serum levels of homocysteine and • Folic acid antagonists, such as methotrexate, inhibit
methylmalonic acid. dihydrofolate reductase and lead to a deficiency of FH4.
• Serum antibodies to intrinsic factor are highly specific for Inhibition of folate metabolism affects all rapidly
pernicious anemia. proliferating tissues, particularly the bone marrow and the
• The diagnosis is confirmed by an outpouring of gastrointestinal tract.
reticulocytes and a rise in hematocrit levels beginning • Many chemotherapeutic drugs used in the treatment of
about 5 days after parenteral administration of vitamin cancer damage DNA or inhibit DNA synthesis through
B12. other mechanisms, and these also cause megaloblastic
• Persons with atrophy and metaplasia of the gastric changes in rapidly dividing cells.
mucosa due to pernicious anemia are at increased risk for
gastric carcinoma. CLINICAL FEATURES:
• With parenteral or high-dose oral vitamin B12, the anemia • General Malnutrition
is cured and the progression of the peripheral neurologic • Cheilosis, Glossitis, Dermatitis
disease can be reversed or at least halted, but the • Folate does not prevent or may even exacerbate
changes in the gastric mucosa and the risk of carcinoma neurologic deficits of Vitamin B12 deficiency.
are unaffected.

c. FOLATE ACID DEFICIENCY ANEMIA


• Dependent on dietary source with 50-200 ug daily
requirements
• A deficiency of folic acid (more properly,
pteroylmonoglutamic acid) results in a megaloblastic
anemia having the same pathologic features as that
caused by vitamin B12 deficiency.
• Among the molecules whose synthesis is dependent on
folates, dTMP is perhaps the most important biologically,
because it is required for DNA synthesis.
• It should be apparent from this discussion that
suppressed synthesis of DNA, the common
denominator of folic acid and vitamin B12 deficiency, is the
immediate cause of megaloblastosis.
THREE MAJOR CAUSES:
1. Decreased intake - chronic alcoholics, elderly, indigent
2. Increased requirements
3. Impaired utilization

• Humans depend on dietary sources for folic acid.


• The richest sources are green vegetables, certain fruits
and animal sources contain lesser amounts. The folic acid
in these foods is largely in the form of
folylpolyglutamates. Although abundant in raw foods,
polyglutamates are sensitive to heat; boiling, steaming, or
frying food for 5 to 10 minutes destroys up to 95% of the
folate content.
• Intestinal conjugases split the polyglutamates into
monoglutamates that are absorbed in the proximal
jejunum.
• During intestinal absorption they are modified to 5-
methyltetrahydrofolate, the transport form of folate. The Figure 12: Megaloblastic Anemia
body’s reserves of folate are relatively modest, and a
deficiency can arise within weeks to months if intake is
inadequate. Decreased intake can result from either a d. IRON DEFICIENCY ANEMIA
nutritionally inadequate diet or impairment of intestinal • Most common nutritional disorder in the world.
absorption. • Results in clinical signs and symptoms that are mostly
• Malabsorption syndromes, such as sprue, may lead to related to inadequate Hgb synthesis.
inadequate folate absorption, as may infiltrative diseases • Common in US, particularly in toddlers, adolescent girls
of the small intestine (e.g., lymphoma). (because of menstruation), women of childbearing age
• Certain drugs, particularly the anticonvulsant phenytoin
and oral contraceptives, interfere with absorption.
• Despite normal intake of folic acid, a relative deficiency
can be encountered when requirements are increased.
Conditions in which this is seen include
1. Pregnancy
2. Infancy
Prepared by: CMED 2B Page 11 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• Disappearance of stainable iron from macrophages using


Prussian Blue stain-smears in the bone marrow.
• Microcytic (small) and hypochromic (pale) cells present.
• Poikilocytosis: small, elongated red cells (pencil cells)

Table 5: Iron Distribution in Healthy Young Adults

• 80% of iron is functional, found in Hgb, myoglobin, and iron


containing enzymes (catalase, cytochromes).
• 15-20% is in the storage pool, in the form of ferritin and
hemosiderin.
• Iron in the body is recycled between the functional and
storage pools. Transported in plasma by an iron-binding
glycoprotein called transferrin, which is synthesized in the
liver.
• Major function is to deliver iron to cells which require it.
• Iron absorption in the duodenum is regulated by hepcidin, Figure 13: Hypochromic microcytic anemia
a small circulating peptide that is synthesized and
released from the liver in response to increases in CLINICAL FEATURES
intrahepatic iron levels. • Non-specific symptoms of anemia.
• Hepcidin inhibits iron transfer from the enterocyte to • Dominating signs and symptoms frequently relate to the
plasma by binding to ferroportin, causing it to be underlying cause.
endocytosed and degraded. • GIT or Gynecologic, Malnutrition, Pregnancy,
• As a result, as hepcidin levels rise, iron becomes trapped Malabsorption
within duodenal cells in the form of mucosal ferritin and is • In severe and long-standing IDA, depletion of iron
lost as these cells slough. containing enzymes in cells throughout the body
• when the body is replete with iron, high hepcidin levels • Depletion from CNS may lead to pica syndrome.
inhibit its absorption into the blood. • Plummer-Vinson Syndrome, a triad of:
• with low body stores of iron, hepcidin levels fall, facilitating o Esophageal webs.
iron absorption. o Microcytic Hypochromic Anemia
• Free iron is highly toxic, there it is sequestered. o Atrophic Glossitis
o Ferritin: a ubiquitous protein-iron complex is found at DIAGNOSIS
highest levels in the liver, spleen, marrow, and • Depressed Hgb and Hct
skeletal muscles bind to iron in storage pools. • Hypochromia, Microcytosis, modest poikilocytosis
o Hemosiderin: partially degraded protein shells of • Serum Iron and Ferritin: LOW
ferritin aggregates, traces amounts principally in • Total Plasma Iron Binding Capacity: HIGH
macrophages. • Low serum iron with increased binding capacity results in
• Plasma Ferritin is derived largely from storage pool, its a transferrin saturation below 15%.
level correlate well with iron stores. So if there is low
plasma ferritin then there is also low iron stores.
ETIOLOGY
e. ANEMIA OF CHRONIC DISEASE
• Dietary insufficiency • Most common cause of anemia among hospitalized patients
• Impaired Absorption (US)
• Increased Requirement (Infants, Pregnant) • Anemia occurs in the setting of persistent systemic
• Chronic Blood Loss inflammation.
• To maintain a normal iron balance, about 1 mg of iron must • Associated with:
be absorbed from the diet every day. o LOW serum iron
• Because only 10% to 15% of ingested iron is absorbed, o DECREASE total iron binding capacity
the daily iron requirement is 7 to 10 mg for adult men and o INCREASE stored iron in tissue macrophages
7 to 20 mg for adult women.
3 categories:
• Chronic blood loss is the most common cause of iron
a. Microbial Infections (Osteomyelitis, Bacterial
deficiency in high income societies.
Endocarditis, Lung Abscess)
PATHOGENESIS
b. Chronic Immune Disorders (Rheumatoid arthritis,
• It produces hypochromic microcytic anemia.
regional enteritis)
• Progressive depletion of the reserves first lowers serum c. Neoplasms - Breast & Lung CA, Hodgkin Lymphoma
iron and transferrin "without anemia".
• Anemia only appears when iron stores are completely PATHOGENESIS
depleted and is accompanied by low serum iron, ferritin, • Certain inflammatory mediators, particularly IL-6 stimulate
and transferrin. Time wherein anemia and pallor can be an increase in hepatic production of hepcidin, which
seen. inhibits iron absorption by
MORPHOLOGY
• Erythropoietin levels are low for the degree of anemia.
• Bone marrow reveals mild to moderate increase in
• Hepcidin can directly or indirectly inhibit erythropoietin
erythroid progenitors.
production.
Prepared by: CMED 2B Page 12 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• Iron sequestration serves to enhance the body's ability to • Initial manifestation depends on cell line affected.
fend off certain organisms that require iron (H. influenzae). • Anemia - progressive weakness, pallor, dyspnea.
• Thrombocytopenia - petechial, ecchymoses, bruises.
CLINICAL LABORATORY FEATURES
• Anemia is usually mild.
• Neutropenia - minor infections, fever, chills.
• Dominant symptoms are those of the underlying disease.
• Normocytic and normochromic, or microcytic and
g. PURE RED CELL APLASIA
hypochromic. • Primary marrow disorder in which only erythroid progenitors
• Increased storage of iron, high serum ferritin, reduced are suppressed.
TIBC. • Severe cases: Completely absent.
• Treatment of the underlying condition corrects the • May occur in association with neoplasms (Thymoma, large
anemia). granular lymphocytic leukemia), drug exposures, autoimmune
disorders, Parvovirus infection.
f. APLASTIC ANEMIA
Parvovirus B19 Infection:
• Refers to a syndrome of chronic primary hematopoietic failure • Normal individuals - Transient Aplasia
and attendance pancytopenia (anemia, neutropenia, and • Moderate to severe - Hemolytic anemias (Aplastic Crises)
thrombocytopenia). • Immunosuppressed (HIV) - Chronic red cell aplasia,
• Majority of patients have autoimmune mechanisms but moderate to severe anemia.
inherited or acquired abnormalities of hematopoietic stem cells
seem to contribute. h. MYELOPHTHISIC ANEMIA
• A form of bone marrow failure in which space-occupying
lesions replace normal marrow element.
• Common cause is metastatic cancer most often carcinomas
from breast, lungs, and prostate.
• Infiltrative process can also occur
• Chronic renal failure, whatever its cause, is almost invariably
associated with an anemia that tends to be
• roughly proportional to the severity of the uremia.

II. POLYCYTHEMIA
• Increase in RBCs
• Denotes an abnormally high red cell count, usually with
corresponding increase in hemoglobin levels.
• Relative: when there is hemoconcentration due to decreased
plasma volume.
• Absolute: increase in total red cell mass
• It also is associated with a condition of unknown etiology
called stress polycythemia, or Gaisböck syndrome.
• Affected individuals are usually males who are hypertensive,
obese, and anxious (“stressed”).
• Absolute polycythemia is primary when it results from an
Table 6: Major Causes of Aplastic Anemia intrinsic abnormality of hematopoietic precursors and
secondary when it stems from the response of red cell
• Specific abnormalities underlying some cases of aplastic progenitors to elevated levels of erythropoietin.
aplasia are as follows. • The most common cause of primary polycythemia is
1. Fanconi anemia polycythemia vera, a myeloproliferative neoplasm
- rare autosomal recessive disorder caused by associated with mutations that lead to erythropoietin-
defects in a multiprotein complex that is required independent growth of red cell progenitors
for DNA repair.
- Marrow hypofunction becomes evident early in
life and is often accompanied by multiple
congenital anomalies, such as hypoplasia of the
kidney and spleen, and bone anomalies,
commonly involving the thumbs or radii.
2. Inherited defects in telomerase are found in 5% to
10% of adult-onset aplastic anemia. Telomerase is
required for cellular immortality and limitless
replication. It might be anticipated, therefore, that
deficits in telomerase activity could result in
premature hematopoietic stem cell exhaustion and
marrow aplasia.
3. Abnormally short telomeres, which are found in the
marrow cells of as many as one-half of those affected
with aplastic anemia. about 65% of cases fall into this
idiopathic category.

CLINICAL FEATURES
• Occurs at any age or sex, insidious onset. Table 7: Pathophysiologic Classification of Polycythemia
Prepared by: CMED 2B Page 13 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

III. BLEEDING DISORDERS (HEMORRHAGIC ⁃ Associated with microvascular bleeding resulting from
collagen defects that weaken walls
DIATHESIS
d. Henoch-Schonlein purpura
• Excess bleeding results from: ⁃ Systemic immune disorder characterized by a purpuric
o Increased fragility of vessels rash, colicky abdominal pain, polyarthralgia, acute
o Platelet deficiency or dysfunction glomerulonephritis.
o Derangement of coagulation
• Normal hemostatic responses involve blood vessel walls, e. Hereditary hemorrhagic telangiectasia (Weber-Osler-
platelets and clotting cascade. Rendu syndrome)
⁃ Characterized by dilated, tortuous blood vessels within
TEST FOR HEMOSTASIS EVALUATION thin walls that bleed readily.
1. Prothrombin time (PT) - assess extrinsic and common
coagulation pathway. f. Perivascular amyloidosis
⁃ Measure the clotting of plasma after addition of exogenous ⁃ Weaken blood vessel walls and cause bleeding.
tissue thromboplastin and calcium ions.
⁃ Prolonged PT result from deficiency or dysfunction of
factor V, VII, X, prothrombin or fibrinogen. B. THROMBOCYTOPENIA
• Reduction in platelet number constitutes an important cause
2. Partial thromboplastin time (PTT) - assess intrinsic and of generalized bleeding.
common coagulation pathways. • Count <100,000 platelets/L is considered thrombocytopenia.
⁃ Clotting of plasma after the addition of kaolin, cephalin and • 20,000 to 50,000 platelets/uL aggravate posttraumatic
calcium ions bleeding. Platelet transfusion is required. In patients with
⁃ Prolongation is due to deficiency or dysfunction of factors cardiac problems and have <70,000 platelets/uL, blood
V, VIII, IX, X, XI, XII, prothrombin, fibrinogen or interfering transfusion is often done during surgery.
antibodies to phospholipids. • <20,000 platelets/uL may be associated with spontaneous
(nontraumatic) bleeding.
3. Platelet counts - electronic particle counter • Bleeding from thrombocytopenia is associated with normal PT
⁃ Reference range: 150-400 x 103/uL I Abnormal counts are and PTT. PT and PTT are for the coagulation pathway.
best confirmed through peripheral blood smear. • Spontaneous bleeding associated with thrombocytopenia
⁃ Platelet clumping can cause spurious thrombocytopenia. most often involves small vessels.
⁃ High counts may be indicative of a myeloproliferative • Common sites are the skin and mucous membranes of the GI
disorder. and GUT.
• Intracranial bleeding is the most feared type, it is a threat to
4. Tests of platelet function include tests for platelet aggregation, many patients with a markedly depressed platelet count.
quantitative tests of von Willebrand factor, bleeding time.
Bleeding time now is replaced by aPTT and PT. CAUSES OF THROMOCYTOPENIA
• Decreased Platelet Production
5. Specialized tests measure levels of specific clotting factors, • Decreased Platelet Survival
fibrinogen, fibrin split products and presence of circulating • Sequestration
anticoagulants. • Dilution

A. BLEEDING DISORDERS CAUSED BY VESSEL WALL 1. CHRONIC IMMUNE THROMBOCYTOPENIC PURPURA


ABNORMALITIES • Caused by auto-antibody mediated destruction of platelets.
• Relatively common disorders that doesn't cause serious • Occur in a variety of predisposing conditions and exposures
bleeding problems. (secondary) or in the absence of risk factors (primary or
• Present with small hemorrhages (petechia or purpura) in idiopathic).
skin or membranes, particularly gingivae. • Secondary Chronic ITP occurs in SLE, HIV infection and B-cell
• Hemorrhages may occur in joints, muscles, subperiosteal neoplasms (CLL).
locations and takes the form of menorrhagia, nosebleeds, • Diagnosis of primary is made after exclusion of secondary
bleeding or hematuria. chronic ITP.
• PT and PTT are usually normal
PATHOGENESIS
CLINICAL CONDITIONS WITH BLEEDING DUE TO VESSEL • Autoantibodies are most often directed against platelet
WALL ABNORMALITIES membrane glycoproteins lIb-Illa or Ib-IX and demonstrate the
plasma and bound to platelet surface in 80% of patient.
a. Infections • Antiplatelet antibodies of IgG class are found in majority the
⁃ Induce petechial and purpuric hemorrhages cases.
⁃ Meningococcemia, septicemia, infective endocarditis,
MORPHOLOGY
rickettsioses
• The principal changes of thrombocytopenic purpura are found
⁃ Mechanisms include vasculitis and DIC.
in the spleen, bone marrow, and blood, but they are not
specific.
b. Drug reactions
• Secondary changes related to the bleeding diathesis may be
⁃ Mediated by deposition of drug induced immune
found anywhere in the body.
complexes in vessel walls leading to hypersensitivity
vasculitis. • The spleen is of normal size. Typically, there is congestion of
the sinusoids and enlargement of the splenic follicles, often
c. Scurvy and Ehler Danlos Syndrome associated with prominent reactive germinal centers.

Prepared by: CMED 2B Page 14 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

• scattered megakaryocytes are found within the sinuses, • CD4 and CXCR4, the receptor and co-receptor for HIV are
possibly representing a mild form of extramedullary found on megakaryocytes.
hematopoiesis driven by elevated levels of thrombopoietin. • HIV infected megakaryocytes are prone to apoptosis and
• The marrow reveals a modestly increased number of impaired ability to produce platelets.
megakaryocytes. Some are apparently immature, with large,
nonlobulated, single nuclei. These findings are not specific but
merely reflect accelerated thrombopoiesis, being found in most 6. THROMBOTIC MICROANGIOPATHIES [THROMBOTIC
forms of thrombocytopenia resulting from increased platelet THROMBOCYTOPENIC PURPURA (TTP) & HEMOLYTIC-
destruction. UREMIC SYNDROME (HUS)]
• bone marrow examination- rule out thrombocytopenias
resulting from marrow failure or other primary marrow • Thrombotic microangiopathy encompasses a spectrum of
disorders. syndromes including TP and HUS.
• The secondary changes relate to hemorrhage, often in the form • Caused by insults that lead to excessive activation of
of petechial bleeds into the skin and mucous membranes. platelets depositing as thrombi in small blood vessels.
• The peripheral blood often reveals abnormally large • In time, the distinguishing features of both have blurred out
platelets (megathrombocytes), which are a sign of accelerated as TP patients lack one feature and US patients have fever
thrombopoiesis. or neurologic dysfunction.

CLINICAL FEATURES a. Thrombotic Thrombocytopenic Purpura (TTP)


• Commonly in adult women <40years old, female to male ratio ⁃ defined as the pentad of fever, thrombocytopenia,
3:1 microangiopathic haemolytic anemia, transient
• Insidious in onset and characterized by bleeding into the skin neurologic deficits and renal failure.
and mucosal surfaces.
• Cutaneous bleeding is in the form of pinpoint hemorrhages b. Hemolytic Uremic Syndrome (HUS)
(petechia) which can give rise to ecchymoses. ⁃ associated with microangiopathic haemolytic anemia
• History of easy bruising, nosebleeds, bleeding from the gums, and thrombocytopenia, prominent of acute renal
hemorrhages into soft tissues from minor trauma. failure and absence of neurologic symptoms, most
• Initially manifest with melena, hematuria or excessive commonly occurring in children.
menstrual flow.
• Subarachnoid hemorrhage and intracerebral hemorrhage are
serious and fatal complications, but rarely occur.

2. ACUTE IMMUNE THROMBOCYTOPENIC PURPURA


• Caused by autoantibodies to platelet but with distinct and
course clinical features.
• A disease of childhood with equal frequency in both sexes.
• Symptoms appear 1-2 weeks after self-limited viral illness
that triggers development of autoantibodies.
• Usually self-limited and resolves spontaneously within 6
months.
• Glucocorticoids are given if thrombocytopenia is severe in
children with viral prodrome, thrombocytopenia persists.

3. DRUG INDUCED THROMBOCYTOPENIA


• Drug can induce thrombocytopenia through direct effects
on platelets and secondary to immunologically mediated
platelet destruction.
• Quinine, Quinidine and Vancomycin binds to platelet
glycoproteins and create antigenic determinants Table 8: Thrombotic Microangiopathiies
recognized by antibodies.
• Thrombocytopenia is also a common consequence of
platelet inhibitory drugs that bind glycoprotein Ilb/Illa.
C. BLEEDING DISORDERS WITH DEFECTIVE
PLATELET FUNCTION
• Several inherited disorders: abnormal platelet function and
4. HEPARIN INDUCED THROMBOCYTOPENIA (HIT) normal platelet count.
TWO TYPES OF HIT:
a. Type I - occurs rapidly after the onset of therapy, resolves THREE PATHOGENICALLY DISTINCT GROUPS:
despite continuation of therapy. Results from a direct 1. Defects of Adhesion - Bernard-Soulier Syndrome
platelet-aggregating effect of heparin. 2. Defects of aggregation - Glanzmann Thrombasthenia
b. Type II - less common but of greater importance, occurring 3. Disorders of platelet secretion (release reaction) -
5-14 days after therapy begins and leads to life threatening Storage Pool Disorders.
venous and arterial thrombosis.
Bernard-Soulier syndrome
- consequences of defective adhesion of platelets to
5. HIV-ASSOCIATED THROMBOCYTOPENIA subendothelial matrix.
• Thrombocytopenia is one of the most common - It is an autosomal recessive disorder caused by an
hematologic manifestations of HIV infections due to inherited deficiency of the platelet membrane glycoprotein
impaired production and increased destruction of platelets. complex Ib–IX. This glycoprotein is a receptor for vWF and
Prepared by: CMED 2B Page 15 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

is essential for normal platelet adhesion to the • More than 20 variants


subendothelial extracellular matrix.
- Affected patients have a variable, often severe, bleeding 1. Reduced quantity of circulating VWF
tendency. a. Type 1-AD
Glanzmann thrombasthenia ⁃ mild to moderate vWF deficiency, 70% cases
- bleeding due to defective platelet aggregation. ⁃ Incomplete penetrance & variable expressivity -
- It is transmitted as an autosomal recessive trait. Mild disease with some missense mutations
- Thrombasthenic platelets fail to aggregate in response to
adenosine diphosphate (ADP), collagen, epinephrine, or b. Type 3 Autosomal Recessive
thrombin because of deficiency or dysfunction of ⁃ Extremely low levels of functional WF with
glycoprotein IIb–IIIa, severe clinical manifestations
- an integrin that participates in “bridge formation” between
platelets by binding fibrinogen. 2. Qualitative defects in VWF
- The associated bleeding tendency is often severe. a. type 2 von Willebrand Disease, with several subtypes
Storage pool disorders ⁃ Type 2A is the most common, 25% of cases
- defective release of certain mediators of platelet activation, ⁃ Autosomal dominant disorder
such as thromboxanes and granule-bound ADP. ⁃ vWF in normal amounts
⁃ Missense mutation with defective multimer
Acquired defects of platelet function assembly
a. Ingestion of Aspirin & other NSAID's
⁃ Aspirin-potent irreversible inhibitor of the enzyme
cyclooxygenase-required for the synthesis of c. HEMOPHILIA A (FACTOR VIII DEFICIENCY)
thromboxane A2 & prostaglandins. • Most common hereditary disease associated with life-
⁃ Play an important role in platelet aggregation & threatening bleeding
release • Mutations in factor VIll, cofactor for factor IX
• X-linked recessive trait-affects mainly males &
b. Uremia homozygous females-inactivation of x chromosome
⁃ Complex pathogenesis of platelet dysfunction bearing normal factor VIl
⁃ Involves defects in adhesion, granule secretion, & • 30% have no family history, caused by new mutations
aggregation. • Clinical severity correlates with level of factor VIll activity:
o severe disease: <1% normal levels
o moderately severe: 2-5% normal levels
D. HEMORRHAGIC DIATHESIS RELATED TO o mild disease: 6-50% normal levels
ABNORMALITIES IN CLOTTING FACTORS
• All symptomatic cases: tendency of easy bruising &
massive hemorrhage after trauma or operative procedure
HEREDITARY DEFICIENCIES • Spontaneous hemorrhages frequently occur -
• Typically affect a single clotting factor hemarthroses (recurrent-progressive deformity-crippling)
• Most common & important inherited deficiency of coagulation • Treatment: Recombinant factor VIII
factors: factor VIll (haemophilia A) and factor IX (haemophilia • 15% with severe hemophilia A develops antibodies-binds
B) and inhibits factor VIll (perceived as foreign)
• Von Willebrand disease - influence both coagulation & platelet
function
• All clotting factor deficiency cause bleeding EXCEPT factor XII d. HEMOPHILIA B (CHRISTMAS DISEASE, FACTOR IX
deficiency
DEFICIENCY)
ACQUIRED DEFICIENCIES • X-linked recessive trait
• Usually involves multiple coagulation factors simultaneously • Variable clinical severity
• Decreased protein synthesis or shortened haft life • 15% cases -factor IX is present but nonfunctional
• PTT prolonged
• Vitamin K deficiency: impaired synthesis of factors, II, VII, IX, X • Specific factor IX assay for diagnosis
& protein C - Severe parenchymal liver disease and DI
• Treatment: Recombinant Factor IX

a. THE FACTOR VIII-vWF COMPLEX


E. DISSEMINATED INTRAVASCULAR
• Two most common inherited disorders of bleeding: hemophilia
A & von Willebrand Disease COAGULATION (DIC)
• Qualitative or quantitative defects • Acute, subacute or chronic thromboembolic disorder
• Factor VIll-essential cofactor of factor IX, made by sinusoidal • Excessive activation of coagulation formation of thrombi in
endothelial cells and Kupffer cells (liver) tubular epithelial cells the microvasculature in the body
(kidney) • Secondary complication of many different disorders like
infections, surgery…
• Sometimes localized to specific organ or tissue
b. VON-WILLEBRAND DISEASE • Consumption of platelets, fibrin & coagulation factors
• Most common inherited bleeding disorders in humans, AD rare • Secondary activation of fibrinolysis
AR • Can present with signs & symptoms relating to tissue
• Bleeding tendency is mild hypoxia & infarction - Myriad Microthrombi
• Most common symptoms: spontaneous bleeding from mucous • Depletion of factors for hemostasis, activation of
membranes (epistaxis), excessive bleeding from wounds, fibrinolysis or both results to haemorrhage
menorrhagia,
• Prolonged BT in the presence of normal platelet count
Prepared by: CMED 2B Page 16 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

ETIOLOGY AND PATHOGENESIS • An unusual form of DIC occurs in association with giant
• Not a primary disease hemangiomas (Kasabach-Merritt syndrome) in which
• Clotting can be initiated by either 2 pathways: thrombi form within the neoplasm because of stasis and
a. Extrinsic pathway: triggered by release of tissue recurrent trauma to fragile blood vessels.
factor (thromboplastin)
b. Intrinsic pathway: activation of factor XII by surface CLINICAL FEATURES
contact with collagen or other negatively charged • Onset can be fuminant due to endotoxic shock or amniotic
substances fluid embolism
• Both results in the generation of thrombin-converts • Insidious & chronic in carcinomatosis, retention of dead
fibrinogen to fibrin fetus
• Clotting in vivo is initiated by exposure of tissue factor, • 50% affected are obstetric patients having complications
which activates factor VII. of pregnancy - disorder is reversible with delivery of fetus
• The most important effect of tissue factor/factor VII • 33% have carcinomatosis
complexes is activation of factor IX, which in turn activates • Few common patterns: hemolytic pnemia, dyspnea,
factor X. cyanosis and respiratory failure, convulsions, coma,
• Activation of factor X leads to the generation of thrombin, oliguria and acute renal failure, sudden or progressive
the central player in clotting. circulatory failure & shock
• At sites where the endothelium is disrupted, thrombin • Acute DIC, associated with obstetric complications or
converts fibrinogen to fibrin; feeds back to activate factors major trauma, is dominated by bleeding diathesis
IX, VIII, and V; stimulates fibrin crosslinking; inhibits • Chronic DIC - cancer patients present with thrombotic
fibrinolysis; and activates platelets, all of which augment complications
the formation of a stable clot.
• To prevent runaway clotting, this process must be sharply DIAGNOSIS
limited to the site of tissue injury. ⁃ Diagnosis is based on clinical observation & laboratory
studies: Fibrinogen levels, platelets, PT, PTT, & FDP
Two major mechanisms trigger DIC
a. Release of tissue factor or thromboplastic substances into PROGNOSIS
the circulation ⁃ Prognosis is highly variable, depends on the underlying
b. Widespread injury to the endothelial cells disorder

• Endothelial injury initiates DIC in several ways. TREATMENT


• Injuries that cause endothelial cell necrosis expose the ⁃ Definitive treatment: remove or treat the inciting cause
subendothelial matrix, leading to the activation of platelets
and the coagulation pathway.
• DIC is most commonly associated with obstetric F. COMPLICATIONS OF TRANSFUSION
complications, malignant neoplasms, sepsis, and major
trauma.
1. Febrile non-hemolytic reaction
• The triggers in these conditions are often multiple and o takes the form of fever and chills, sometimes with mild
interrelated. dyspnea and occurs within 6 hours of a transfusion of
red cells or platelets, the most commonly
Possible consequences of DIC: encountered
1. Widespread deposition of fibrin within the microcirculation
ischemia of more severely affected or more vulnerable 2. Allergic Reactions
organs o Severe, potentially fatal allergic reactions may occur
⁃ Microangiopathic hemolytic anemia-fragmentation of when blood products containing certain antigens are
red cells given to sensitized recipients
2. Consumption of platelets & clotting factors & activation of o IgA deficiency, triggered by IgG antibodies
plasminogen-hemorrhagic diathesis o Urticarcial allergic reactions - triggered by presence
of an allergen in donated blood that is recognized by
MORPHOLOGY IgE antibodies
• Thrombi most often found in: brain, heart, k idneys,
adrenals, spleen & liver 3. Hemolytic Reactions
• Kidneys: small thrombi in glomeruli-reactive swelling of o Caused by preexisting formed IgM antibodies against
endothelial cells, severe cases-microinfarcts, bilateral donor red cells that fix complement
cortical necrosis o Delayed hemolytic reactions are caused by
• Lungs: numerous fibrin thrombi in alveolar capillaries- antibodies that recognized red cell antigens that the
pulmonary edema & fibrin exudation-"hyaline membranes" recipient was sensitized to previously, through prior
- reminiscent of ARDS blood transfusion
• Sheehan postpartum pituitary necrosis - DIC complicating
labor & delivery 4. Transfusion Related Acute Lung Injury (TRALI)
• Toxemia of pregnancy - placenta exhibits widespread o Severe, frequently fatal complication in which factors
microthrombi and premature atrophy of cytotrophoblast& in a transfused blood product trigger the activation of
syncytiotrophoblast. neutrophils in the lung vasculature
• Giant hemangiomas are thrombi due to stasis & recurrent
trauma to fragile blood vessels 5. Infectious Complications
• In meningococcemia, fibrin thrombi within the o Bacterial infections are caused by skin flora,
microcirculation of the adrenal cortex are the probable indicating that the contamination occurred at the time
basis for the massive adrenal hemorrhages seen in that the product is taken from the donor
Waterhouse-Friderichsen syndrome. o Advances in donor screening, selection and
infectious disease testing have dramatically
Prepared by: CMED 2B Page 17 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

decreased the incidence of viral transmission by 9. Pica, a clinical presentation for Fe deficiency
blood products anemia, is
a. Itchiness
b. ED
IV. Reference c. Desire to eat weird things
d. A small woodland creature
Dr Refuerzo PowerPoint Presentation. RBC and Bleeding
Disorders. January 4, 2022
10. Which lab investigations would you order if you
suspect Fe deficiency anemia? (check all that apply)
Kumar, V., Abas, A., Aster, J. Robbins & Cotran Pathologic Basis of
a. CBC
Disease (10th ed). Philadelphia, USA. Elsevier. (2019).
b. Blood smear
c. Serum iron
V. Test Yourself d. Serum ferritin
e. TIBC
f. All of the above
1. Which of the following is NOT a cause of microcytic
anemia? 11. Where is beta-thalassemia most common? (check
a. Thalassemia all that apply)
b. Anemia of chronic disease a. West Africa
c. Iron deficiency anemia b. Mediterranean
d. Pancytopenia c. Arabian Peninsula
e. Lead poisoning d. South East Asia
e. Canada
2. The lab reports for a patient with low mean cell volume
show high serum ferritin and low total iron binding 12. What is the difference between beta-thalassemia
capacity. What is the most likely cause for major and beta-thalassemia minor?
this patient’s anemia? a. Homozygous vs heterozygous
a. Fe deficiency b. Acute vs chronic
b. Anemia secondary to inflammation c. Legal drinking age
c. Thalassemia
d. Hemoglobinopathy 13. Heinz bodies are made of:
a. Excess gamma chains
3. Fe is absorbed in the b. Excess alpha chains
a. Stomach c. Excess beta chains
b. Duodenum d. Excess ketchup
c. Jejunum
d. Ileum 14. Beta-thalassemia, unlike alpha-thalassemia, presents at
approximately 6 months of age.
4. Where is most nonheme iron found in the body? a. True
a. Bound to IF b. False
b. Bound to transferrin
c. Free in plasma 15. Which would you expect to see on a blood smear
d. Stored in liver for beta-thalassemia? (select all that apply)
a. Heinz bodies
5. Select the following that enhance Fe absorption b. Multinucleated neutrophils
(select all that apply) c. Target cells
a. Citric acid d. Hypochromic microcytic cells
b. Polyphenols (tea) e. Hyperchromic microcytic cells
c. Phytate (bran)
d. Calcium 16. What is the treatment for beta-thalassemia minor?
e. Ascorbic acid a. Blood transfusions
b. Iron chelation
6. What is the most important test for Fe stores? c. Bone marrow transplant
a. Serum iron d. None of the above
b. TIBC
c. Serum ferritin 17. Decreased or stopped production of alpha-globin chains
results in HbH (4 gamma chains together) and Hb Barts
7. Which of the following is not an etiology of Fe (4 beta chains together)
deficiency anemia? a. True
a. Chronic blood loss b. False
b. Increased requirement
c. Infection 18. On a CBC for alpha-thalassemia, you would see anemia
d. Malabsorption and reticulocytosis. On the blood smear, you would see
e. Decreased intake Heinz bodies, hypochromic microcytic cells, and
occasional target cells. Select the others that you would
8. TIBC increases in iron deficiency anemia because see increase:
a. Inflammatory response to deficiency a. LDH
b. Compensation by other factors b. Unconjugated bilirubin
c. Ability to absorb increases c. Conjugated bilirubin

Prepared by: CMED 2B Page 18 of 20


Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

d. Urine urobilinogen b. No neurological symptoms in folic acid


e. Urine hemosiderin c. Muscle wasting
d. Dizziness
19. Aplastic anemia can be acquired (more common)
and inherited. What are some of the ways it can be 29. Folic acid therapy can cause sickle cell anemia
acquired? a. True
a. Postviral infection b. False
b. Pregnancy
30. Hydroxyurea increases hemoglobin production and
c. Ionizing radiation
decreases reticuolocyte cells.
d. Drugs and chemicals
e. Idiopathic a. True
f. All of above b. False

20. Aside from the gradual onset signs of anemia, what other 31. Which test can be used to detect hemolytic anemia?
clinical presentations would you see with aplastic a. Coombs test
anemia? b. Genetic testing
a. Koilonychias, “spoon nails” c. Peripheral blood smear
b. Associated thrombocytopenia, e.g., history of d. Schilling Test
bleeding from the gums
c. Neutropenia, e.g., repeated bacterial infections 32. Which anemia is classified as not being able to use iron
d. Purpura properly to synthesize hemoglobin because of an
e. Pica inherited cause.
a. Iron deficiency anemia
21. How would you diagnose aplastic anemia?
b. Hypochromic anemia
a. Blood smear
b. Bone marrow biopsy c. Aplastic anemia
c. Spleen biopsy
d. CBC 33. What is the lifespan of RBC?
e. Liver biopsy a. 120 days
b. 100 days
22. Select treatment options for aplastic anemia c. 200 days
a. IV equine ATG d. 80 days
b. Bone marrow transplant
c. Splenectomy 34. The hexose monophosphate pathway activity increases
d. Immune suppression the RBC source of
a. Glucose and lactic acid
b. 2,3-BPG and methemoglobin
23. Causes spurious decrease in MCV c. NADPH and reduced gluthathione
a. Cryofibrinogen
d. ATP and other purine metabolites
b. hyperglycemia
c. autoagglutination
d. high WBC ct 35. Which single feature of normal RBC’s is most responsible
e. reduced red cell deformability for limiting their lifespan?
a. Loss of mitochondria
24. When the entire CBC is suppressed due to either b. Increased flexibility of the cell membrane
anemia, infection or hemorrhage is called? c. Reduction of Hb iron
a. Erythroplasia d. Loss of nucleus
b. Thrombocytopenia
c. Pancytopenia
d. Leukopenia Answer Key

25. Red Cell indices: Determination of relative size of RBC –


82-98fL
a. MCH 1. D 11. B,C,D 21. B 31. A
b. MCV
c. MCHC 2. B 12. A 22. A,B,D 32. B
3. B 13. B 23. A 33. A
26. Measurement of average weight of Hgb/RBC 4. B 14. A 24. C 34. C
a. MCH 5. A 15. A, C, D 25. B 35. D
b. MCV 6. C 16. D 26. B
c. MCHC 7. C 17. B 27. C
8. C 18. A,B,D,E 28. B
27. Evaluation of RBC saturation with Hgb 9. C 19. F 29. B
a. MCV
b. MCH
10. D 20. BCD 30. A
c. MCHC

28. Vitamin b 12 and folic have the similar adverse effects


but what separates one from the other?
a. Glossitis
Prepared by: CMED 2B Page 19 of 20
Pathology Lecture

(013) RBC and Bleeding Disorders


DRA. REFUERZO| 01/04/22

VI. Appendix

Prepared by: CMED 2B Page 20 of 20

You might also like