Download as pptx, pdf, or txt
Download as pptx, pdf, or txt
You are on page 1of 40

Jones Ofori-Amoah ( PhD MPhil MPH BSc

DipHSM DipHRM)

Department of Pharmacology and Toxicology, School of Pharmacy, UHAS.

First ask
yourself:
what is the
worst that
can happen?
Then prepare
to accept it.
Toxicology
Then proceed 1
OBJECTIVES

• Mechanisms of cell damage and cell death


• Biological Protective mechanisms
• Effects of toxicants on the kidney and liver
• Toxic responses to the blood

2
CELLULAR TOXICITY
MECHANISMS

3
Mechanisms of cell damage and cell death

• Toxic compounds damage cells in several ways.


• Cellular injury leads to a complex sequence of
events

Eventual response may be:


• reversible injury
• irreversible, leading to the death of the cell
(necrosis)

4
Cell damage/death:
The process leading to cell death can be divided
into 3 events:

Primary: those that result in initial damage


Secondary: cellular changes which follow from the
initial damage
Tertiary: these are the observable and final changes

5
Primary events
• Many toxic compounds are toxic following metabolism to
reactive intermediates (metabolites)

• The reactive metabolites may then initiate one or more


primary events.

• The major primary events are:


• Lipid peroxidation
• Oxidative stress
• Covalent binding to macromolecules
• Changes in thiol status
• Enzyme inhibition
• Ischaemia
6
Lipid peroxidation:
 Free radical formation by –
 homolytic cleavage of a covalent bond in a molecule
 addition of an electron
 abstraction of a H atom by another radical

 Have an unpaired electron on C, N, S or O, i.e. a reactive sp.


that can react with cellular components such as
polyunsaturated fatty acids found in cell membranes

 Examples: carbon tetrachloride – a hepatotoxin


destruction of B-cells by alloxan
destruction of nerve terminal by 6-hydroxydopamine

7
Oxidative stress:
• Production of active oxygen sp.
 May lead to redox cycling with electrons being donated to
yield superoxide.

Changes in thiol status:


• The cell can be overwhelmed by excessive RIs
• RIs can also oxidise GSH and thiol groups in proteins and change
the thiol status of the cell

GSH plays a key role in the protection of the cell by:


- direct chemical reaction with reactive intermediates
- a GSH transferase mediated reaction
8
Enzyme inhibition:
Can be a common consequence of exposure to toxic compounds
e.g. inhibition of cytochrome aa3 by cyanide – leads to blockage of
cellular respiration

Ischaemia:
• Cessation or reduction of the supply of blood containing O2 and
nutrients such as glucose, leads to tissue damage and cell death
if prolonged
• Hypoxia and anoxia may be specific effects leading to cell death.

Covalent binding to macromolecules:


• RI can bind to macromolecules and proteins covalently.
• There is correlation between this binding and tissue damage.
9
Secondary events
Changes in membrane structure and permeability – could be a
consequence of lipid peroxidation
Changes in cytoskeleton
Mitochondrial damage
Inhibition of mitochondrial function
Depletion of ATP and other co-factors
DNA damage
Lysosomal destabilization – toxic compounds may rupture lysosomes
and destroy the cell.
Changes in intracellular Ca2+ concentration ⇒⇒ leads to:
• Alterations in cytoskleton
• Activation of phospholipases which leads to breakdown of membrane
phospholipids
• Ca2+-activated proteases
• Ca2+-activated endonucleases – cleave DNA into fragments
10
Tertiary Changes
• Steatosis/fatty changes
• Hydropic degeneration
• Blebbing of the plasma membrane

Morphological changes
Two distinct forms of morphological change before cell death include:
 Apoptosis: orderly process of cell shrinkage, condensation, blebbing
and phagocytosis.
 Necrosis: disorderly process involving swelling, and the rupture of
membranes, dissolution of organized structures, loss of homeostasis
and ATP.
11
Apoptosis
 Occurs as part of the normal maintenance and renewal of
tissues
• Stimulated by toxic chemicals
 Triggers usually cell injury particularly involving DNA
• Cells attempt to repair injury which might fail, leading to a
mutated cell which can undergo clonal expansion.
• the cell may die by apoptosis to prevent clonal expansion of a
potentially initiated cell.
• The exposure to a sufficient dose of a chemical can initiate a
process of gene activation leading to the synthesis of specific
proteins that drive the apoptotic machinery.
• Gene products involved include fos, myc jun, bax, p53, caspases

12
Necrosis
 Occurs due to irreversible damage followed by a
series of degenerative changes such as hydrolysis of
cellular components and denaturation of proteins.

 Necrosis is associated with:


• massive increase in cell volume
• swelling of the mitochondria & ER
• appearance of vacuoles and the accumulation of fluid
• ultimate rupture of the cell membrane and spillage of the
cytoplasmic contents into the tissue surroundings

13
Biological Protective mechanisms
Biological systems possess a number of mechanisms
for protection against toxic foreign compounds

• Metabolic transformation to more polar


metabolites readily excreted is a method of
detoxification
• e.g. conjugation and sulphation of paracetamol

14
Glutathione (GSH)
• Probably the most important protective mechanism.
• GSH is found in most cells and at a relatively high concentration
in the liver (5mM)
• It has a nucleophilic thiol group and can detoxify substances in
one of 3 ways

-conjugation catalysed by glutathione transferase


-chemical reaction with a reactive metabolite to form a
conjugate
-donation of a proton or hydrogen atom to reactive metabolites
or free radicals respectively

15
Superoxide dismutase (SD)
Free radical or other reactive intermediates
may donate electrons to O2 form:

• Active O2 such as superoxide anion radical


which can cause cellular damage

SD removes superoxide to produce H2O2  catalase 


water + O2

16
Vitamins and Amino acids

• Apart from GSH, lipid radicals and other radicals may


be removed by a number of endogenous compounds;
e.g. Vitamin E, Vitamin K

• Others include Cysteine, Ascorbate.


These compounds act as alternative hydrogen donors in
preference to the allylic H atoms of unsaturated lipids .

17
TARGET ORGAN
TOXICITY

18
Target Organ/Tissue Toxicity
Although any organ or tissue may be a target for a toxic compound,
such compounds often damage specific organs or tissues.

Reasons:
• Blood supply; e.g. kidney vrs skin
• The presence of particular enzymes or biochemical pathway
• The function or position of the organ
• The vulnerability to disruption or degree of specialization
• The ability of the organ to repair damage
• The presence of particular uptake systems
• The ability to metabolize the compound, and the balance of
toxification and detoxification
• Binding to particular macromolecules; e.g. melanin of the eye
19
Toxic damage to the Liver
• One of the portals of entry to the tissues in the body
• Exposed to many potentially toxic substances by the GI tract from
the
-diet
-food additives
-contaminants
-drugs

• By virtue of its position, structure, function and biochemistry,


the liver is vulnerable to damage from drugs.

• Liver damage in man is however less common as only 9% of


adverse drug reactions affect the liver.
20
The liver is a target organ for toxic substances for 4
main reasons
• The liver metabolizes many xenobiotics but metabolism
may not always result in detoxification. E.g. CCl4 & Paracetamol

• The liver also has an extensive role in intermediary metabolism


and synthesis. Interference with endogenous metabolic pathways
may lead to toxic effects.

• The liver receives 25% of the cardiac output. The blood


supply ensures that the liver is exposed to relatively high
concentrations of toxic substances absorbed from the GIT

• The secretion of bile may also be a factor. Biliary excretion


of foreign compounds leading to high concentrations especially
if saturation occurs; e.g. frusemide
21
Types of liver injury
• Fatty liver (steatosis): accumulation of triglycerides in
hepatocytes
-triglyceride synthesis occurs in the liver, fatty liver – a direct
result in the reduced synthesis of apolipoprotein responsible for
transporting triglycerides out of the liver. E.g. alcohol, tetracycline

• Cholestatic damage: interference with the biliary system can


lead to bile stasis. e.g. chlorpromazine, rifampicin

• Cirrhosis: A chronic lesion resulting from repeated injury


and subsequent repair. May result from hepatocyte or cholestatic
damage; e.g CCl4 and alcohol

22
Types of liver damage cont’d
Vascular lesions:
• Some substances may be directly cytotoxic to the
hepatic sinusoids and the endothelial cells, causing
damage and occlusion of the lumen.
• Blood flow in the liver is reduced leading to ischaemia
and necrosis; eg monocrotaline

Liver tumours:
Both benign and malignant tumours have been
associated with the use of oral contraceptives and
exposure to aflatoxins
23
Detection of liver damage
• Bilirubin levels increase in liver damage
• Plasma albumin levels also decrease in liver damage
• Liver enzymes (such as AST, ALT, ALP) which are raised
several fold within 24 hours of liver damage, may also
be measured in plasma
• Urinary excretion of conjugated bilirubin
• Light and electron microscopy
• Liver weight/body weight ratio

24
Paracetamol toxicity
• Paracetamol, widely used as an analgesic and
antipyretic
o Relatively safe in therapeutic doses
• In over-dosage, paracetamol causes hepatic
necrosis; sometimes accompanied by renal damage
and failure
• Now common for overdose of paracetamol to be
taken for suicidal intent, abortive purposes, etc.
 E.g. in the UK, around 200 deaths a year from overdose of
paracetamol

25
The metabolites of paracetamol

26
Proposed metabolic activation of paracetamol to a toxic, reactive
intermediate N-acetyl-p-benzoquinone imine (NAPQI). NAPQI can react
with GSH to form a conjugate or with tissue proteins.

27
Treatment of paracetamol over-dosage

• N-Acetylcysteine: acts by several mechanisms


-it promotes the synthesis of GSH utilized in
the conjugation of the reactive metabolite NAPQI

-It stimulates the synthesis of GSH used in the


protection of protein thiols

-it may relieve the saturation of sulphate conjugation


which occurs during paracetamol over-dosage

- it may be directly involved in the reduction of NAPQI


28
• NAC originally thought to be effective only when administered 10-12 hours
after an over-dosage.
• But now found to be effective even after 15 hours or more after over-
dosage.
• Arguably, majority of the metabolic activation and covalent binding to
protein would have taken place within 12 hours and it appears unlikely
that it will be reacting with NAPQI.

• NAC is suspected to protect liver cells against the subsequent changes


which occur after the reaction of the RI with cellular constituents.

• Methionine: not effective when given at such later times since synthesis of
GSH from methionine is inhibited or compromised by paracetamol toxicity.
• Inhibition of thiol containing enzymes involved in the synthetic pathway
likely to be the cause.

29
Toxicity to the kidneys
The kidney is a target for toxic agents for the following reasons:

• Blood through the kidneys represent 25% of the Cardiac output


despite the kidneys forming 1% of body mass – High exposure to
toxins

• Concentration ability of the kidneys: many substances are


re-absorbed after glomerular filtration
• concn of toxicants in the tubular lumen can be extremely
high; e.g. sulphonamides causing crystalluria.

30
Kidney as a target organ for toxicants
• Active transport: compounds which are actively transported
from the blood into the tubular fluid may accumulate in the
proximal tubular cells especially at high concns where saturation
of the transport system occurs.

• Concentration to which tubular cells are exposed may be higher


than in the blood stream. E.g. cephaloridine, which causes
proximal tubular damage.

• The kidney does not contain much cyt. P450 enzymes, but there
is sufficient to activate certain substances; e.g. Chloroform &
paracetamol.

31
Detection of kidney damage
• Variety of ways of detecting kidney damage ranging from
simple qualitative tests to more complex biochemical assays

 Simple tests:
• Urine volume
• pH
• SG measurements
• Presence of cells or protein in urine
• Kidney wt/body wt ratio

• Clinically overt pathological damage detected by light or


Electron microscopy.

32
 Damage can also be detected by measurement of a variety of
urinary constituents.
• Damage to tubular cells results in the leakage of enzymes such as -
glutamyltransferase & N-acetylglucosaminidase into the tubular fluid.
 Damage may also reflect in altered renal tubular function
leading to:
- the excretion of glucose and amino acids
 measurement of urea & creatinine (raised) will also indicate renal
dysfunction
 Commonest measurement is the measurement of Urea or
blood urea nitrogen (BUN)
 Clearance of the polysaccharide inulin is a better indicator
of G.F., as it is less affected by protein metabolism
33
Sites for Nephrotoxicity
Difficult to define precisely the specific site of toxicity as
several tissue components may be affected:

This is because:
• Cell damage may occur due to interruption of one or
several of the many essential cellular functions rather than
from interaction with specific cellular type.

• Certain kidney cells may be particularly sensitive because


they are exposed to very high conc. of the compound
leading to a non-specific effect.
 E.g. many compounds affect the proximal tubule.
34
• Proximal convoluted tubule: site of absorption of glucose and
amino acids; sensitive to metals such as chromium

• Pars recta of the proximal tubule: Main site of secretion of


organic compounds, contains the highest concn of enzymes;
sensitive to mercury, cephaloridine

• Loop of Henle: site of damage from chronic exposure to


analgesics; e.g. Aspirin, phenacetin

• Distal convoluted tubule: involved in acidification of urine;


affected by amphotericin

• Collecting ducts: Insensitive to most toxicants; site of damage


from outdated tetracyclines
35
Glomerulus: Primary site of damage by several
compounds, also sensitive to immunological damage

Basement membrane: main barrier to filtrate. Toxicity


can lead to change in permeability of this membrane
causing loss of proteins; damaged by puromycin

36
Toxicity to kidneys:
 Aminoglycosides
 Important cause of renal damage in humans
• They are carbohydrates with glycosidic linkages to side chains
containing various amino acids.
 E.g. Gentamycin has 5 amino groups – cationic (achieves a concn
in the renal cortex 2-5 times more than in the blood).
• There appears to be a correlation between the number of
ionisable groups & nephrotoxicity
• It is believed that the cationic aminoglycosides bind to anionic
phospholipids of the proximal tubule.
 Bound aminoglycoside is engulfed by endocytosis and stored in
lysosomes
 Lysosomes eventually rupture and release hydrolytic enzymes to
mediate the cellular damage. 37
 Cephalosporins
E.g. Cephaloridine causes kidney damage in humans
 Acute doses cause proximal tubular necrosis, it accumulates
in the cortex.
• An organic anion, but has a cationic pyridyl group and is
actively secreted by the cationic system into the tubular
lumen.
• There is also an anionic transport system for re-uptake into
the tubular cell which is more active
 the overall effect is accumulation
• Movement of cephaloridine via facilitated diffusion out of the
tubular cell into the lumen also seems to be restricted hence
intracellular concn remains high.
38
Blood as a Target Tissue
 Almost all foreign compounds are distributed via the
bloodstream
• Components of blood are exposed at least initially to high
concns of foreign compounds

 Damage to and destruction of blood cells result in a variety of


events leading to reduced ability to carry oxygen to the tissues
if red cells are involved. E.g. Aniline, dapsone, nitrobenzene
 Their reactive intermediates (hydroxylamines) are unstable and can be
further oxidized to reactive products in the presence of oxygen in the
red cell and damage Hb.

 Hb may then be oxidized to methHb and be unable to carry O2


39
• Irreversible damage to Hb can occur as a result of oxidation of
thiol groups in the protein.

• Proteins become denaturated leading to haemolysis of red


cells. E.g. Phenylhydrazine

• The bone marrow is a target for toxins as the cells are rapidly
dividing;
 e.g. Benzene, chloramphenicol will attack dividing cells in
the bone marrow to induce aplastic anaemia.

40

You might also like