You have 3 free guides left 😟
Unlock your guides
You have 3 free guides left 😟
Unlock your guides

Metabolism is a crucial process in toxicology, determining how our bodies handle harmful substances. It involves two main phases: modify chemicals, while help eliminate them. Understanding these processes is key to grasping how toxins affect us.

The body's metabolic system, particularly the liver, plays a vital role in breaking down and removing toxins. Factors like , sex, and diet can influence metabolism, impacting how individuals respond to harmful substances. This knowledge is essential for predicting and managing toxic effects.

Phases of metabolism

  • Metabolism plays a crucial role in determining the fate and of xenobiotics in the body
  • reactions are divided into two main phases: Phase I and Phase II reactions
  • These reactions modify the chemical structure of xenobiotics to facilitate their elimination from the body

Phase I reactions

Top images from around the web for Phase I reactions
Top images from around the web for Phase I reactions
  • Involve oxidation, reduction, or hydrolysis of the xenobiotic
  • Catalyzed by enzymes such as (CYP) monooxygenases and flavin-containing monooxygenases (FMOs)
  • Examples include hydroxylation of aromatic compounds (benzene) and oxidation of alcohols (ethanol)
  • Phase I reactions often result in the formation of reactive intermediates that can be more toxic than the parent compound
  • These reactions typically increase the polarity of the xenobiotic, making it more water-soluble

Phase II reactions

  • Involve conjugation of the xenobiotic or its Phase I metabolite with endogenous molecules
  • Catalyzed by transferase enzymes such as UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs)
  • Examples include glucuronidation of morphine and sulfation of acetaminophen
  • Phase II reactions generally increase the molecular weight and polarity of the xenobiotic, facilitating its
  • Conjugation reactions typically detoxify the xenobiotic, although some exceptions exist (acetaminophen-glucuronide)

Cytochrome P450 system

  • The cytochrome P450 (CYP) system is a superfamily of heme-containing enzymes that play a central role in xenobiotic metabolism
  • CYP enzymes are predominantly expressed in the liver but are also found in extrahepatic tissues (intestine, lung, brain)
  • These enzymes catalyze a wide range of Phase I reactions, including oxidation, reduction, and hydrolysis

Structure and function

  • CYP enzymes are membrane-bound proteins located in the endoplasmic reticulum
  • They contain a heme group that binds molecular oxygen and a substrate-binding site that determines substrate specificity
  • The catalytic cycle involves the activation of oxygen, leading to the insertion of one oxygen atom into the substrate and the formation of water
  • Examples of CYP-mediated reactions include the hydroxylation of steroids (testosterone) and the epoxidation of polycyclic aromatic hydrocarbons (benzo[a]pyrene)

Genetic polymorphisms

  • CYP enzymes exhibit significant genetic variability, resulting in interindividual differences in drug metabolism and toxicity
  • Polymorphisms can lead to altered enzyme activity, ranging from complete loss of function to increased activity
  • Examples include CYP2D6 polymorphisms affecting the metabolism of antidepressants (fluoxetine) and CYP2C19 polymorphisms influencing the response to proton pump inhibitors (omeprazole)
  • Genetic testing can help predict an individual's metabolic capacity and guide personalized drug therapy

Induction and inhibition

  • The activity of CYP enzymes can be modulated by various factors, including drugs, environmental pollutants, and dietary components
  • involves an increase in the expression or activity of CYP enzymes, leading to enhanced metabolism of substrates
  • Examples of CYP inducers include rifampicin (CYP3A4) and polycyclic aromatic hydrocarbons (CYP1A1)
  • results in decreased metabolism of substrates, potentially leading to drug-drug interactions and toxicity
  • Examples of CYP inhibitors include grapefruit juice (CYP3A4) and fluconazole (CYP2C9)

Conjugation reactions

  • Conjugation reactions are Phase II biotransformation reactions that involve the covalent attachment of endogenous molecules to xenobiotics or their Phase I metabolites
  • These reactions are catalyzed by transferase enzymes and require the presence of cofactors (UDP-glucuronic acid, 3'-phosphoadenosine-5'-phosphosulfate)
  • Conjugation reactions generally increase the polarity and molecular weight of the xenobiotic, facilitating its excretion via urine or bile

Glucuronidation

  • Catalyzed by UDP-glucuronosyltransferases (UGTs) in the endoplasmic reticulum
  • Involves the transfer of glucuronic acid from UDP-glucuronic acid to the xenobiotic or its Phase I metabolite
  • Examples include the glucuronidation of morphine and acetaminophen
  • Glucuronidation typically detoxifies xenobiotics, although some exceptions exist (morphine-6-glucuronide)

Sulfation

  • Catalyzed by sulfotransferases (SULTs) in the cytosol
  • Involves the transfer of a sulfonate group from 3'-phosphoadenosine-5'-phosphosulfate (PAPS) to the xenobiotic or its Phase I metabolite
  • Examples include the sulfation of steroid hormones (estradiol) and phenolic compounds (acetaminophen)
  • Sulfation can lead to the formation of reactive intermediates (N-sulfooxy-2-acetylaminofluorene)

Acetylation

  • Catalyzed by N-acetyltransferases (NATs) in the cytosol
  • Involves the transfer of an acetyl group from acetyl-coenzyme A to the amino group of the xenobiotic or its Phase I metabolite
  • Examples include the acetylation of isoniazid and sulfonamides
  • Acetylation exhibits genetic polymorphism, leading to slow and fast acetylator phenotypes

Methylation

  • Catalyzed by methyltransferases in the cytosol
  • Involves the transfer of a methyl group from S-adenosylmethionine (SAM) to the xenobiotic or its Phase I metabolite
  • Examples include the methylation of catecholamines (dopamine) and arsenic
  • Methylation can lead to the formation of toxic metabolites (monomethylarsonic acid)

Glutathione conjugation

  • Catalyzed by glutathione S-transferases (GSTs) in the cytosol
  • Involves the conjugation of the tripeptide glutathione to the xenobiotic or its Phase I metabolite
  • Examples include the conjugation of electrophilic compounds (aflatoxin B1) and reactive oxygen species
  • Glutathione conjugation is an important pathway, although some conjugates can be toxic (S-(1,2-dichlorovinyl)-L-cysteine)

Factors affecting metabolism

  • Various factors can influence the rate and extent of xenobiotic metabolism, leading to interindividual variability in toxicity and drug response
  • These factors include age, sex, nutritional status, disease states, and drug interactions
  • Understanding the impact of these factors is crucial for personalized medicine and risk assessment

Age and development

  • Metabolic capacity changes throughout the lifespan, with significant differences between neonates, children, adults, and the elderly
  • Neonates have immature enzyme systems, leading to reduced metabolism of certain drugs (theophylline)
  • Elderly individuals may have decreased liver mass and blood flow, resulting in altered drug clearance (benzodiazepines)
  • Developmental changes in enzyme expression can influence the susceptibility to toxicants (chlorpyrifos)

Sex differences

  • Sex-related differences in xenobiotic metabolism can be attributed to hormonal influences and genetic factors
  • Some CYP enzymes (CYP3A4) exhibit higher activity in females, while others (CYP1A2) have higher activity in males
  • Sex differences in metabolism can affect drug efficacy and toxicity (zolpidem)
  • Hormonal fluctuations during the menstrual cycle and pregnancy can alter metabolic capacity

Nutritional status

  • Diet and nutritional status can modulate the activity of drug-metabolizing enzymes
  • High-protein diets can induce CYP enzymes (CYP3A4), while malnutrition can lead to decreased enzyme activity
  • Specific dietary components can inhibit or induce enzyme activity (grapefruit juice, cruciferous vegetables)
  • Nutritional deficiencies (vitamin B6, iron) can impair the function of certain enzymes (cysteine conjugate β-lyase)

Disease states

  • Various disease states can alter the expression and activity of drug-metabolizing enzymes
  • Liver diseases (cirrhosis, hepatitis) can reduce the metabolic capacity of the liver
  • Inflammation and infection can downregulate CYP enzymes through the action of cytokines (interleukin-6)
  • Genetic diseases (Gilbert's syndrome) can lead to impaired conjugation reactions (glucuronidation)

Drug interactions

  • Drug-drug interactions can occur when one drug alters the metabolism of another drug
  • Enzyme induction by one drug can lead to increased metabolism and reduced efficacy of another drug (rifampicin and oral contraceptives)
  • Enzyme inhibition by one drug can result in decreased metabolism and increased toxicity of another drug (ketoconazole and midazolam)
  • Herbal supplements and dietary components can also interact with drugs by modulating enzyme activity (St. John's wort, grapefruit juice)

Metabolic activation vs detoxification

  • Xenobiotic metabolism can result in either detoxification or of the parent compound
  • Detoxification involves the conversion of the xenobiotic into less toxic or inactive metabolites, facilitating their elimination from the body
  • Bioactivation, or metabolic activation, involves the formation of reactive intermediates that can be more toxic than the parent compound
  • The balance between detoxification and bioactivation determines the ultimate toxicity of a xenobiotic

Bioactivation of toxicants

  • Many toxicants require metabolic activation to exert their toxic effects
  • Examples include the bioactivation of benzo[a]pyrene to diol epoxides that form DNA adducts and the activation of acetaminophen to N-acetyl-p-benzoquinone imine (NAPQI) that causes liver toxicity
  • Bioactivation often involves Phase I reactions catalyzed by CYP enzymes, leading to the formation of electrophilic intermediates
  • These reactive intermediates can bind to cellular macromolecules (DNA, proteins) and disrupt cellular function

Detoxification pathways

  • Detoxification pathways convert xenobiotics into less toxic or inactive metabolites that can be readily excreted from the body
  • Examples include the glucuronidation of morphine and the sulfation of acetaminophen
  • Detoxification often involves Phase II conjugation reactions that increase the polarity and water solubility of the xenobiotic
  • Glutathione conjugation is an important detoxification pathway for electrophilic compounds and reactive oxygen species

Balance and consequences

  • The balance between bioactivation and detoxification determines the net toxicity of a xenobiotic
  • Factors that influence this balance include the relative activities of Phase I and Phase II enzymes, the availability of cofactors, and genetic polymorphisms
  • When bioactivation overwhelms detoxification, the accumulation of reactive intermediates can lead to cellular damage and toxicity (acetaminophen overdose)
  • Interindividual differences in the balance between bioactivation and detoxification can contribute to variability in susceptibility to toxicants (aflatoxin B1)

Toxicokinetics of metabolism

  • Toxicokinetics describes the , , metabolism, and excretion (ADME) of xenobiotics in the body
  • Metabolism plays a crucial role in determining the fate and toxicity of xenobiotics
  • Understanding the toxicokinetics of metabolism is essential for predicting the exposure, bioavailability, and clearance of xenobiotics

Absorption and distribution

  • Absorption refers to the entry of a xenobiotic into the systemic circulation from the site of exposure (oral, dermal, inhalation)
  • Factors influencing absorption include the physicochemical properties of the xenobiotic (lipophilicity, molecular weight) and the route of exposure
  • Distribution describes the reversible transfer of the xenobiotic from the systemic circulation to various tissues and organs
  • The extent of distribution depends on factors such as plasma protein binding, tissue affinity, and the presence of tissue-specific transporters

Metabolic clearance

  • Metabolic clearance refers to the rate at which a xenobiotic is irreversibly removed from the body through metabolism
  • Clearance is determined by the activity of drug-metabolizing enzymes and the blood flow to the metabolizing organs (liver, kidneys)
  • High metabolic clearance can lead to rapid elimination of the xenobiotic, while low clearance can result in accumulation and prolonged exposure
  • Interindividual differences in metabolic clearance can contribute to variability in drug response and toxicity

Elimination half-life

  • The elimination half-life is the time required for the plasma concentration of a xenobiotic to decrease by 50% during the elimination phase
  • The half-life is determined by the clearance and the volume of distribution of the xenobiotic
  • Xenobiotics with short half-lives are rapidly eliminated from the body, while those with long half-lives can accumulate with repeated exposure
  • Knowledge of the half-life is important for determining dosing intervals and assessing the risk of toxicity

Bioavailability and bioaccumulation

  • Bioavailability refers to the fraction of the administered dose that reaches the systemic circulation unchanged
  • Factors influencing bioavailability include the extent of absorption, , and presystemic elimination
  • Bioaccumulation occurs when the rate of uptake of a xenobiotic exceeds the rate of elimination, leading to an increase in the body burden over time
  • Xenobiotics with high lipophilicity and low metabolic clearance are more likely to bioaccumulate (PCBs, dioxins)

Organ-specific metabolism

  • While the liver is the primary site of xenobiotic metabolism, other organs and tissues also possess metabolic capabilities
  • Organ-specific metabolism can influence the local toxicity of xenobiotics and contribute to the overall metabolic fate of the compound
  • Understanding the role of extrahepatic metabolism is important for predicting tissue-specific toxicity and drug-drug interactions

Liver as primary site

  • The liver is the principal organ responsible for xenobiotic metabolism due to its high expression of drug-metabolizing enzymes and its central role in blood circulation
  • Hepatocytes, the main cell type in the liver, express a wide range of Phase I and Phase II enzymes (CYP enzymes, UGTs, GSTs)
  • The liver's high metabolic capacity and blood flow make it a major site for first-pass metabolism and systemic clearance of xenobiotics
  • Liver-specific toxicity can occur when reactive metabolites are formed or when detoxification pathways are overwhelmed (acetaminophen-induced hepatotoxicity)

Extrahepatic metabolism

  • Extrahepatic tissues, such as the intestine, lung, kidney, and skin, also express drug-metabolizing enzymes
  • The intestine is a major site of first-pass metabolism for orally administered xenobiotics due to the presence of CYP enzymes and UGTs in enterocytes
  • The lung is exposed to inhaled xenobiotics and can metabolize compounds through CYP enzymes and phase II reactions (benzene, naphthalene)
  • The kidney plays a role in the metabolism and excretion of xenobiotics and can be a target for tissue-specific toxicity (chloroform)

Blood-brain barrier

  • The blood-brain barrier (BBB) is a selectively permeable barrier that regulates the entry of xenobiotics into the central nervous system (CNS)
  • The BBB is formed by tight junctions between endothelial cells and the presence of efflux transporters (P-glycoprotein)
  • Xenobiotics that are lipophilic and have low molecular weight can cross the BBB more readily (ethanol, caffeine)
  • Metabolism of xenobiotics by CYP enzymes and phase II reactions in the brain can influence their CNS effects and toxicity (nicotine, polycyclic aromatic hydrocarbons)

Placental transfer

  • The placenta is a selective barrier that regulates the transfer of xenobiotics from the maternal circulation to the fetal compartment
  • Placental transfer depends on factors such as the physicochemical properties of the xenobiotic, the stage of pregnancy, and the presence of placental transporters
  • Some xenobiotics can cross the placenta and expose the developing fetus to potential toxicity (thalidomide, alcohol)
  • The placenta expresses drug-metabolizing enzymes (CYP enzymes, UGTs) that can influence the extent of fetal exposure to xenobiotics and their metabolites

Metabolic disorders and toxicity

  • Metabolic disorders, whether inborn or acquired, can alter the body's ability to metabolize xenobiotics and endogenous compounds
  • These disorders can lead to the accumulation of toxic metabolites or the impaired detoxification of xenobiotics
  • Understanding the impact of metabolic disorders on toxicity is important for risk assessment and patient management

Inborn errors of metabolism

  • Inborn errors of metabolism are genetic disorders that affect the synthesis, degradation, or transport of specific molecules
  • Examples include phenylketonuria (PKU), which impairs the metabolism of phenylalanine, and galactosemia, which affects the metabolism of galactose
  • These
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.
Glossary
Glossary