Vous êtes sur la page 1sur 119

What is Toxicology?

• Toxicology is the study of the adverse effects of chemical,


physical, or biological agents on people, animals, and the
environment.

• The word “toxicology” is derived from the Latinized form of the


Greek word “toxikon” which means "arrow poison”.

• Toxicologists are scientists trained to investigate, interpret, and


communicate the nature of those effects.
Nomenclature of Classification

• Toxins-naturally poisons
– Venoms
– Exotoxins (diphtheria and tetrodotoxin)
– Plants (atropa belladonna)
– Gases (hydrogen sulfide)
• Toxicants-synthetic poisons
– Industrial solvents
– Medications
– Heavy metals*
Exposome

• The “exposome” is defined as the measure of all the exposures of an


individual in a lifetime and how those exposures relate to health.

• An individual’s lifetime exposure history begins before birth and includes


insults from environmental and occupational sources.

• Understanding how exposures from our environment, diet, lifestyle, etc.


interact with our own unique characteristics (i.e., genetics, epigenetics and
physiology) to impact our health is how the exposome is articulated.

• https://www.cdc.gov/niosh/topics/exposome/
Regulatory Toxicology
Toxic Substances Control Act

• TSCA stands for the Toxic Substances Control Act of 1976.

• Provides EPA with authority to require reporting, record-keeping and testing


requirements, and restrictions relating to chemical substances and/or mixtures.

• Certain substances are generally excluded from TSCA, including, among others,
food, drugs, cosmetics and pesticides.

• TSCA addresses the production, importation, use, and disposal of specific chemicals
including polychlorinated biphenyls (PCBs), asbestos, radon and lead-based paint.

• http://www2.epa.gov/laws-regulations/summary-toxic-substances-control-act
REGULATORY AGENCIES

Food and Drug Administration (FDA)


- Drugs, Food, Food additives, Cosmetics, Medical Devices

Environmental Protection Agency (EPA)


- Pesticides, Industrial Chemicals, Hazardous Waste, Pollutants
in air, water and soil

Occupational Safety and Health Administration


(OSHA)
- Chemicals in workplace
Toxicology: Integrated Summary

• The Food and Drug Administration (FDA) requires an


integrated summary of the preclinical toxicology studies of a
novel drug be submitted with all Investigational New Drug (IND)
applications.

• Pre-Clinical Studies Required for IND applications:


– Acute Toxicity
– Subchronic Toxicity
– Genetic Toxicity
– Safety Pharmacology
– Toxicokinetics
Regulatory Risk Assessment
• Importance in setting allowable levels of
chemicals in air, water, workplace by EPA and
OSHA
– TLV: threshold limit value, guideline of American
Conference of Governmental Industrial Hygienists
– PEL: permissable exposure level, OSHA regulation
– RfD: reference dose, EPA maximum acceptable oral
dose
• Challenge of extrapolation from:
– Animals to humans
– Experimental dose (LOEL, lowest observed effect
level in animals) to low dose in humans
International Council for Harmonization (ICH)
Risk v. Risk Assessment

• Risk
– the probability of an adverse outcome

• Risk assessment
– the systematic scientific characterization of potential adverse
health effects resulting from exposure to hazardous agents of
situations.
Objectives of Risk Assessment

• Identify and evaluate chemical hazards


• Determine factors that influence exposure to
chemical hazards
Components of Risk Assessment

• Hazard identification
• Dose-response assessment
• Time-response assessment
• Exposure assessment (route and source)
• Risk characterization
Hazard identification
• Organ/tissue sites of toxicity
– hepatic, renal, cardiovascular
• Type of Pathology
– carcinogenic, teratogenic, cytotoxic
• Structure of chemical
• Mechanism of toxicity
– Important for extrapolation such as animal to human
• Study Type
– Computational, in-vitro, preclinical animal study,
clinical study, epidemiological study, case report
Hazard Identification and Risk Prediction

• Structure
• Modeling
• C-statistic
• Machine learning
• In vitro assays
• In vivo studies
Risk Assessment:
Define Specific Exposure Circumstances

• Intensity (concentration)
• Duration (length of time)
• Acute/Chronic
• Recent/Remote
Measuring and Reporting Toxicity

• LD50: Lethal dose in 50% of animals


• LOAEL: Lowest Observable Adverse Effects Level
• NOAEL: No Observable Adverse Effect Level
• ED50: Effective dose in 50% of animals/humans
• TI: Therapeutic Index for animals
• TD50: Toxic dose in 50% percent of human subjects
• TR: Therapeutic Ratio for humans
• MOS: Margin of Safety
Characterization of Chemical Hazards
By Severity of Effects
• Dose response relationships
• No Observed Adverse Effect Level (NOAEL)
• Lowest Observed Adverse Effect Level (LOAEL)
Exposure Levels

• Dose (amount absorbed)


• Body burden (accumulated dose)
• Threshold of effect (level of exposure resulting in
measurable or observable effects)
Acute Reference Dose

• The acute reference dose (ARD) is an estimate of a daily oral


exposure for an acute duration (24 hours or less) that is likely to
be without an appreciable risk of deleterious effects during a
lifetime.

• The ARD can be derived from a NOAEL, LOAEL, or benchmark


dose, with uncertainty factors generally applied to reflect
limitations of the data used.

• Generally used in EPA's non-cancer health assessments.


Assessing and Defining
Exposure by Duration
• Acute (single acute exposure; < 96 hours)
• Subacute (repeated or continuous exposure for
4 to 30 days)
• Subchronic (repeated or continuous exposure
for 30 to 90 days)
• Chronic (repeated or continuous exposure for >
3 months)
• Lifetime (Exposome)
Define Hazard By Onset and Duration of Effects

• Acute Toxicity
– Acute toxicity is used to describe effects which appear promptly, or
within 24 hours of exposure
• Chronic Toxicity
– Chronic toxicity is the delayed effect of exposure to a hazardous
chemical substance.
– It is measured in experimental conditions after three months of either
continuous or occasional exposure.
• High acute toxicity does not necessarily equal high chronic
toxicity.
• Low acute toxicity does not necessarily equal low chronic
toxicity.
Benefit/Risk Analysis
• Therapeutic benefit v. side effects (adverse effects, toxic
effects)

• Adverse effect
– An adverse effect may be termed a "side effect", when judged to
be secondary to a main or therapeutic effect.

• The therapeutic index (TI) (also referred to


as therapeutic ratio) is a comparison of the amount of
a therapeutic agent that causes the therapeutic effect to the
amount that causes toxicity.
Types of Biological Markers
• Markers of exposure:
– Internal dose
– Biologically effective dose
• Markers of effect:
– Biological effects (measurable)
– Altered cellular structures (biopsy studies)
• Markers of susceptibility:
– Genetic polymorphisms
– Diseases that can alter metabolism of chemicals (e.g., liver
failure)
Interpretation of Biological Marker Data

• Relevance based on:


A) Sensitivity
B) Specificity
Toxicity
Classification
Characterization of Risk
Absolute risk based on LD50 (mg/kg)
Toxic Substance LD50 mg/kg
Ethanol 10,000

Morphine 1,500

Nicotine 1

Tubocurare 0.5

Dioxin 0.001

Botulinum toxin 0.00001


Risk Characterization
• Risk based on selectivity
– Probability of efficacy vs probability of toxicity
– Measured by therapeutic index or relative risk
Risk Characterization
• Risk based on selectivity
– Probability of efficacy vs probability of toxicity
– Measured by therapeutic index or relative risk
Dose Dependent Toxicity
LD50 = Lethal Dose 50% or median lethal dose. Amount of the
substance required to kill 50% of the test population.

LD50 may vary greatly due to factors such as the animal


species tested and mode of administration.

Chocolate
Essentially harmless to humans, toxic to many animals e.g.,
dogs

Venom
LD50 results may be misleading due to the physiological
differences between mice, rats, and humans e.g., venomous
snakes have venom adapted to incapacitate mice.
Dose Dependent Toxicity
Margin of Safety

• The goal of drug discovery is to find effective and safe


treatments for disease. Because as previously mentioned all
drugs are potential toxicants, safety is relative to the intended
use of the drug.

• The difference between the minimal effective dose and the


minimum dose that causes toxic side effects is called the margin
of safety.
Risk Characterization

Therapeutic index (TI)= LD50/ED50


The lower TI, the smaller the margin of safety, e.g. digoxin, 2.0
Risk Characterization Issues
• Animal to
human and
human to
human
extrapolation

• Low dose
extrapolation
Allometric Scaling
• Empirical approach where the exchange of drug dose is based
on normalization of dose to body surface area.
Defining Exposure Circumstances

• In utero
• Childhood
• Adulthood
• Comorbid with illness
Age and Risk Characterization
• Younger individuals are more sensitive to toxic
substances than older individuals.
• The difference can be primarily attributed to
differences in metabolism; specially to differences in
the functioning of the liver.
• Lead absorption rate is four to five times greater in
young individuals than in adults
• Cadmium absorption rate is 20 times greater than in
adults.
Risk Characterization of Special Populations

• Phenotype (thin v. obese)


• Genotype
– The differences in genetics makeup both within species
and between species markedly influence the disposition
and the metabolism of toxic substances.
• The liver and the kidney function are
– Important organs for detoxifying and removing toxic
substances.
– Liver or kidney disease can enhance the toxic effects of
substances normally detoxified by these organs.
Sex and Risk Characterization

• The difference in toxicity between genders seems to be


primarily influence by the sex hormones (estrogen and
testosterone), which can affect metabolism.
• Parathion (organophosphate) is metabolized more rapidly in
females resulting in a higher concentration of the more toxic
intermediate, paraoxon.
• Male rats are 10 times more susceptible to liver damage than
female rats as a result of chronic oral exposure to DDT; due to
DDT acts directly on the testes to altering the neuroendocrine
function in male rats.
Mechanisms of Toxic Action
Mechanism of Toxicity

• Binding to protein (receptors, enzymes)


• Binding to DNA
• Oxidative stress
– GSH depletion, formation of O2 radicals, lipid
peroxidation
• Increased intracellular free calcium ion
Electrophilic Alkylating Agents
• An electrophile is a chemical that is attracted to electrons.

• Electrophiles are positively charged or neutral species.

• These chemicals are attracted to the electron rich centers of nucleophiles.

• Methylation is the most common type of alkylation, being associated with


the transfer of a methyl group. Biological methylation is distinct from
electrophilic alkylation which transfers of long chain carbon groups to
nucleophilic centers of cellular macromolecules.

• Electrophilic alkylating agents are often very toxic, due to their ability to
alkylate DNA; alkylated DNA does not uncoil properly.

• This results in cytotoxicity and inhibition of cell growth and initiation of


apoptosis. In addition carcinogenic mutations can be introduced resulting in
higher incidence of cancer after exposure.
CYP 450 Mediated Epoxide Formation
Oxidative Stress
Calcium Influx
Types of Cytotoxic
Mechanisms
• Necrosis (Gr. : death)
– Swelling, membrane lysis, inflammation

• Apoptosis (Gr. : falling off)


– Programmed cell death
– Signal transduction pathway causing
characteristic DNA fragmentation, membrane
‘blebbing’
Drug Induced Liver Injury (DILI)
Clinical Manifestations of
Exposure to Hepatoxic Agents
• Fatty liver or steatosis (due to inhibition of very low density lipoprotein
synthesis and accumulation of triglycerides)

• Necrosis (focal or zonal; zone 3 most susceptible to injury)

• Cirrhosis (accumulation of collagen in liver; fibrosis)

• Hepatic encephalopathy (associated with cirrhosis)

• Cholestasis (e.g., impaired bile formation and flow which can be due to
several factors including disruption of canicular contractility by chemicals
such as cyclosporin)

• Cancer
Biomarkers of Hepatic Dysfunction:
• Aspartate aminotransferase (AST) increased by necrosis of
hepatocytes
• Alanine aminotransferase (ALT) increased by necrosis of
hepatocytes
• Bilirubin (product of hemoglobin degradation; total (indirect)
and conjugated (direct)
• Alkaline phosphatase (dephosphorylation of compounds.
• Hy’s Law (Dr. Hyman Zimmerman):
– The drug causes hepatocellular injury with jaundice, generally defined as an
elevated ALT and AST by 3-fold or greater above the upper limit of
normal.
– Among subjects showing such aminotransferase elevations, they also have
elevation of their serum total bilirubin of greater than 2× the upper
limit of normal, without findings of impaired bile formation (defined as
serum alkaline phosphatase activity less than 2× the upper limit of normal).
In vitro Chromosome Aberration Assay
• Identifies clastogenic agents (clastogens) which cause large scale structural damage
(chromosome/chromatid aberrations) to chromosomes leading to leading to
sections of the chromosome being deleted, added, or rearranged.

• Clastogenic agents interfere with cell cycle and mitotic spindle apparatus function.

• This assay can also detect aneugens (numerical changes in number of


chromosomes)

• Primary cells (human peripheral blood lymphocytes) or cell lines (Chinese Hamster
Ovary cells a.k.a. CHO) are used

• Assay has good sensitivity but poor specificity (positive for non-carcinogens)

• Susceptible to extreme cell culture conditions (pH, osmolality, cytotoxicity)


Micronucleus test (MNT)

• Micronucleus is the name given to the small nucleus that forms whenever a
chromosome or a fragment of a chromosome is not incorporated into one of the
daughter nuclei during cell division.

• The micronucleus test (MNT) is used to determine if a compound is genotoxic by


evaluating the presence of micronuclei.

• Micronuclei may contain chromosome fragments produced from DNA breakage


(clastogens) or whole chromosomes produced by disruption of the mitotic apparatus
(aneugens).

• Approximately 24 hours after the last dose, bone marrow or peripheral blood is
collected to determine the frequency of micronucleated polychromatic
erythrocytes (MN-PCEs) or micronucleated reticulocytes (MN-RETs),
respectively.

• A positive outcome is characterized by a statistically significant, dose-dependent


increase in MN-PCEs or MN-RETs that exceeds historical control limits.
Micronuclei
Bacterial Reverse Mutation Assay
(Ames Test)

• Identify agents mutagens that cause point mutations (additions,


subtractions, substitutions) in DNA

• Salmonella typhimurium and E. coli were engineered to require


histidine and tryptophan respectively for growth.
– At least five strains of bacteria are used. These should include four strains of S.
typhimurium (TA1535; TA1537 or TA97a or TA97; TA98; and TA100)

• Exposure to mutagens causes reversion wherein the bacteria can


once again synthesize amino acids: this reversion can be detected as
colony formation in appropriate medium.

• The assay exhibits good concordance for carcinogens


In vitro Mouse Lymphoma Assay (MLA)

• The mouse lymphoma TK assay (MLA) is part of an in vitro battery of tests designed to
predict risk assessment prior to in vivo testing.
– L5178Y mouse lymphoma tk ( +/- ) cells
– Measuring resistance to the lethal nucleoside analogue triflurothymidine (TFT).
– Detects mutagenic and clastogenic events induced by TFT

• The test has the potential to detect mutagenic and clastogenic events at the thymidine
kinase (tk) locus of L5178Y mouse lymphoma tk ( +/- ) cells by measuring resistance to
the lethal nucleoside analogue triflurothymidine (TFT).

• When added to selective medium containing TFT, wild-type tk ( +/- ) cells die, but TFT
cannot be incorporated into the DNA of mutant tk ( -/- ) cells, which survive
– Large colonies are indicative of gene mutations.
– Small colonies are indicative of large deletions (chromosomal mutations).
– Dose response mutant frequency is expressed as the number of mutants per 10(6) viable
cells.
Assessment of Carcinogenic Risk

• In vitro studies to test for potential


carcinogenicity and mechanism
• Lifetime rodent studies with at least three
exposure levels for determination of D-R curve
• Estimation of human exposure

• See EPA Guidelines for Carcinogenic Risk Assessment 2005


https://www.epa.gov/risk/guidelines-carcinogen-risk-assessment
Mechanisms of Carcinogenicity
• Genotoxic: alter DNA causing
– activation of proto-oncogene
• Polycyclic aromatic hydrocarbins (PAHs) mutate p21ras (small
GTPase) by depurination

– inhibition of tumor suppressor gene


• Aflatoxin causes G:C to T:A mutation in p53 (enhancer of DNA
repair and inhibitor of cell replication)

• Nongenotoxic (epigenetic): increase DNA


replication, decrease cell death, increase clonal
expansion of transformed cell
Rodent Carcinogenicity
Studies
• Use of maximally tolerated dose (MTD)
– <10% decrease in body weight and no effects
predicted to shorten lifespan, based on subchronic
study
• Problem of determining dose with acceptably
low risk eg. less than 1/10,000
– How to extrapolate the D-R curve from small rodent
sample, such as 50 animals, with measured tumor
incidence down to dose in large human population for
acceptably low tumor risk
– Linear extrapolation to zero dose with zero risk as
default method and appropriate for mutagens;
nonlinear approach must be justifiable
Immunotoxicology

• Sites of Action of Immunotoxicants:


– Proteins (hapten mediated hypersensitvity reactions)
– Immunoglobulins
– Lymphocytes (activation or suppression of innate and
acquired immune cell proliferation)
• Mitogens (agents that induce mitosis) promote lymphocyte
proliferation
Innate Immunity Assays:
Macrophage Phagocytosis Assay
Macrophage Phagocytosis Assays
– Measure of primary innate immune
response.
– For fluorescent beads we simply
count beads in the macrophages
– To measure the chromated sheep
erythrocytes taken up by
macrophages we use a scintillation
counter.
– Same assay in vivo using radio
labeled particulates is called the
Reticuloendothelial System Assay

Natural Killer Cell Assay


– Measures ability of natural killer cells to
target labeled tumor cells.
TDAR Assay of Humoral Immunity
Local Lymph Node Assay
• Used to measure the
induction phase of the
DTH response to contact
and respiratory allergens
– Antigen is topically
applied to the ear for 3
days.
– On day 5 the animal is
injected with
radiolabeled thymidine
(i.e., tritiated thymidine)
– 5 hours later animal
euthanized and lymph
nodes analyzed for
levels of tritiated
thymidine in
proliferating cells.
Footpad Swelling Assay

• The foot pad swelling assay


is a preclinical in vivo test
used to measure the
elicitation phase of the
delayed-type hypersensitivity
response or DTR.
– Animal injected with antigen
subcutaneously near base
of tail to sensitize it.
– One week later, one foot
pad is injected with antigen
and the other with saline
– Swelling is then measured
the following day.
Examples of Toxic Substances
• Drugs:
– Acetaminophen
• Pesticides
• Gases
– Carbon monoxide
– Hydrogen sulfide
• Aromatic hydrocarbons
– dioxins (PAHs)
– benzene, benzopyrene (PAH)
• Metals
Acetaminophen
• Toxic acetaminophen dosages.
• In adults, acute ingestion of more than 150
mg/kg or 12 g of acetaminophen is
considered a toxic dose and poses a high risk
of liver damage.
• In children, acute ingestion of 250 mg/kg or
more poses significant risk for
acetaminophen-induced hepatotoxicity.
Acetaminophen Toxicity
Acetaminophen Toxicity
• Phase 1 (0.5-24 hours after ingestion)
• Patients may be asymptomatic or report anorexia, nausea
or vomiting, and malaise
• Physical examination may reveal pallor, diaphoresis,
malaise, and fatigue

• Phase 2 (18-72 h after ingestion)


• Patients develop right upper quadrant abdominal pain,
anorexia, nausea, and vomiting
• Right upper quadrant tenderness may be present
• Tachycardia and hypotension may indicate volume losses
Acetaminophen Toxicity
• Phase 3: Hepatic phase (72-96 h after ingestion)
• Patients have continued nausea and vomiting,
abdominal pain, and a tender hepatic edge
• Hepatic necrosis and dysfunction may manifest
as jaundice, coagulopathy, hypoglycemia, and hepatic
encephalopathy
• Acute renal failure develops in some critically ill patients
• Death from multiorgan failure may occur

• Phase 4: Recovery phase (4 d to 3 wk after ingestion)


• Patients who survive critical illness in phase 3 have
complete resolution of symptoms and complete
resolution of organ failure
Toxicology of Pesticides

• Source of toxicology data


– Registration required with EPA
– Data submission includes environmental
impact, food residue, animal toxicity
(carcinogenic and reproductive risk)
• Persons at highest risk
– Agricultural workers (applicators, nursery
workers)
Mechanisms of Toxicity

• Inhibition of acetylcholinesterase
– Organophosphates
– Carbamates
• Inhibition of mitochondrial electron transport
chain function
– Paraquat
– Rotenone
• Prevent the closure of sodium channels
– Organochlorines (Lindane, dieldrin, DDT)
– Pyrethriods
Classes of Insecticides
and their Toxicity

• Organochlorines, e.g., DDT


– Na channel activation
– Environmental persistence and toxicity to avian
and aquatic species
– Long half-life in humans
– Concern about ‘endocrine disruption’ based on
hormonal effects in animal models
• Current debate about bisphenol A, a component of
certain plastic products
Special Populations

• Children are particularly vulnerable to the effects of


organochlorines due to their surface area to body
weight ratio and that the Blood-Brain Barrier is not
completed formed.

• Seizures have been reported in children exposed


organochlorines used to treat head lice.
Organochlorine Insecticides

• DDT (dichlorodiphenyltrichloroethane)
• Methoxychlor
• Lindane
• Aldrin
• Dieldrin
• Endosulfan
Organophosphates

• Used as insecticides and chemical warfare agents.

• Organophosphorus pesticides can be absorbed by all routes,


including inhalation, ingestion, and dermal absorption.

• Organophosphates are esters and thus, these compounds are


often used as plasticizers and as additives in high pressure
lubricants

• Thiophosphoryl compounds (i.e., those bearing the P=S


functionality) such as malathion and parathion are much less
toxic to humans than the related phosphoryl derivatives and for
this reason these are often used as insecticides.
Ginger Jake

• During prohibition, people consumed a


homemade alcoholic drink called
Jamaican ginger that was contaminated
with triorthocresyl phosphate (TOCP).

• More than 20,000 people were affected


by a condition called "Ginger Jake
paralysis."

• Although TOCP does not significantly


inhibit the activity of acetylcholinesterase
it does inhibit Neuropathy Target Esterase
(NTE).
Herbicides 1
• Roundup
– glyphosate, the active ingredient, inhibits enzyme
needed for synthesis of three aromatic amino
acids that are required for growth
– weed resistance has lead to increased amounts of
use
– current concerns about human carcinogenicity
with differing opinions by WHO International
Agency for Research on Cancer, European Food
Safety Authority, FAO/WHO Meeting on Pesticide
Residues
Herbicides 2
• 2,4-Dichlorophenoxyacetic acid
– Mimic of plant growth hormone auxin
– Classified as human possible carcinogen by WHO
IARC
– One component of product referred to as Agent
Orange (used as defoliant in Vietnam and Laos
1961-1970, Operation Ranch Hand)
• the other related component 2,4,5-T now banned due
to its toxicity and that of contaminant TCDD generated
in manufacturing process
The Dioxin TCDD
Contaminant of the Herbicide Agent Orange

Tumorigenic in animal models; classified as known


human carcinogen
Toxicity of Dioxins

• Increased gene transcription via Aryl


Hydrocarbon (AH) receptor
–CYP1A1
• Cancer in some animal species
– Marked species variation in LD50
• Chloracne in humans
– Soft tissue sarcoma?
Dioxins increase transcription of genes such as CYP1A1
Chloracne
Occupational Dermatoses
Carcinogenicity of
Aromatic Hydrocarbons
• Benzene: leukemia
• Benzopyrene: lung cancer
• Oxidation by CYP450s and epoxide hydrolase
generates mutagenic metabolites

benzopyrene epoxide diol


Deaths Resulting from Unintentional Carbon Monoxide
Poisoning, by Month and Year — National Vital Statistics
System, United States, 2010–2015
Effect of CO on O2 Dissociation Curve

• The binding of carbon


monoxide to hemoglobin not
only displaces oxygen, but it
also shifts the oxygen
dissociation curve to the left
resulting in less oxygen being
released to tissues with low
PO2.

• Thus during exposure to CO,


the percent saturation of
hemoglobin with O2 remains
high when it should be low.
CO Effects ATP Dependent Processes

• CO disrupts delivery of oxygen which


is necessary for ATP dependent
processes.

• This results in disruption of the ATP


dependent processes including
glutamate/glutamine cycle.

• This results in bioaccumulaiton of


glutamate at presynaptic terminals
which promotes excitotoxicity and
initiation of apoptosis.
T1

T2
Hydrogen Sulfide - H2S

• Colorless gas with a characteristic odor of rotten eggs


– Decay of sulfur-containing organic compounds
– Release from natural gases, volcanic gases, hot springs,
sewage
– Found in petroleum
– Most commonly a by-product of industrial processes
– 92% of exposures occur in natural gas refineries
Mechanism of Action

• Cellular hypoxia:
– Inhibits mitochondrial cytochrome-c oxidase
– Leads to histotoxic hypoxia and metabolic acidosis
Clinical Effects
Concentration
Effect
(ppm)
0.13 - 30 rotten egg odor
30 odor becomes sickeningly sweet
100-150 nose and eye irritation, gas eye
150 olfactory nerve paralysis
keratocojunctivitis, coughing, sore
250-500 throat, chest tightness, pulmonary
edema
headache, loss of consciousness,
500-1000
coma, convulsions
> 1000 Respiratory paralysis, death
Toxic Effects of Metal
Compounds
• Environmental, occupational, medical
exposures
• Risk of toxicity dependent on:
– molecular form of metal
– exposure route
– duration and dose
– host genotype and phenotype
Carcinogenic Metals
Inorganic Arsenic Skin, lung
methylated form
Beryllium Lung

Chromium Lung
hexavalent form
Nickel Nasal, laryngeal,
subsulfide lung
Mees Lines

• Mees' lines or Aldrich-Mees'


lines are horizontal lines of
discoloration which occur on the
nails of the fingers and toes
after an episode of poisoning
with arsenic or thallium or other
heavy metals. They can also
appear if the subject is suffering
from renal failure.

• These are typically white bands


traversing the width of the nail.
With growth of the nail, these
are displaced upward on the
nail and eventually disappear
when trimmed.
Nodular Hyperkeratoses from Arsenic-
Contaminated Well Water
Blackfoot Disease
Thallium

• Acute exposure
– Abdominal cramps
– Constipation
– Nausea and vomiting
• Chronic exposure
– Alopecia
– Hyperkeratosis
– Neuropathy
Lead Toxicology

• Toxic effects of lead described during Roman


Empire with mining, smelting and use in water
pipelines and other purposes
• Environmental contamination in U.S.
throughout the 20th century due to:
– Use in paints until 1976
– Use as tetraethyl lead in gasoline from 1922-1995
(banned in 1990)
Lead

• Toxic forms or species


– Inorganic
– Organic (tetraethyl lead)
• Metabolism
– Demethylation of organic forms
• Mechanism of Toxicity
– Displaces calcium
– Interacts with sulfhydryl groups and disrupts heme biosynthesis
Blue Gingival Lines of Lead Poisoning
Bone Lead
Lowest Observed Effect Level of
Lead in Children
dALA dehydratase <5 ug/dl
IQ and hearing 5-10 ug/dl

Nerve conduction 20 ug/dl

GI colic, anemia, 50-100 ug/dl


nephropathy,
encephalopathy
Death 130 ug/dl
IQ and Blood Lead in 7-yr-old
Children (The Port Pirie Cohort)
NEJM 327:1279, 1992
Blood Pb in Children
Pirkle et al. JAMA 272:285,1994

1976-1980

1988-1994
% Childhood Blood Pb > 10 ug/dL
by Ethnicity and Survey Period
Change in Flint MI Water Source and
Increase in Elevated Blood Pb

Hanna-Attisha et al Am J Pub Hlth 106:283, 2016 (posted)


Geochemical Cycle of Hg
Mercury

• Toxic forms or species


– Elemental
– Inorganic
– Organic
• Metabolism
– All forms are metabolized to inorganic divalent mercury
which is excreted
• Mechanism of Toxicity
– Has a high affinity for sulfhydryl groups
– Binds to microsomes and mitochondria
Toxic Effects of
Elemental and Organic Mercury

• Elemental mercury vapor


– Neuropsychiatric effects
– Tremors
• Organic mercurials
– Paresthesias
– Ataxia
– Deafness and tunnel vision
– CNS development
Victims of Organic Hg Exposure
1956: Minimata disease described in Japan.
1959: Research identified cause as alkyl Hg release into
Minamata Bay, local source of drinking water, by Chisso Corp.
More Hg Concerns

• Use of Hg in research
– See case report on fatality
https://en.wikipedia.org/wiki/Karen_Wetterhahn

• Thimerosal in vaccines as preservative


– Use ended in 2003 except for multi-dose vials
of influenza vaccine, no link to autism
• Hg vapor released from dental amalgams
• Hg in fish and its consumption during
pregnancy
2001 National Academy of Sciences Report on Developmental
Methylmercury Neurotoxicity led to:
• FDA advisory on fish consumption: avoid 1st 4 species below in pregnant and
breast-feeding women and in children
• EPA reduction in safe exposure level: = 1 can tuna per week
• 2014 FDA and EPA recommendation for minimum fish consumption per week

NEJM 347:1735, 2002


For data on exposure reports and cases of
fatalities each year in the U.S., see annual
reports of AAPCC.

Clinical Toxicology, December 2016,


2015 Annual Report of the American Association
of Poison Control Centers
Toxic Exposure Surveillance System.
http://www.aapcc.org/annual-reports/

POSTED in folder for next class.

Vous aimerez peut-être aussi