SlideShare a Scribd company logo
1 of 233
Genetic Toxicology
Dr R B Cope BVSc BSc(Hon   1) PhD   cGLPCP DABT ERT
Quick Review of Relevant
    Basic Biology
Definitions

 Genotoxic:

 Non-genotoxic:

 Direct-acting mutagen:

 Indirect-acting mutagen:

 Pro-mutagen:

 Proximate mutagen:
Definitions

 Ultimate mutagen:

 Alkylating agent:

 Radiomimetic:

 Non-radiomimetic:

 DNA macrolesion

 DNA microlesion
Direct-acting mutagens

 By definition, these are highly DNA reactive agents that
  do not require prior metabolism for direct DNA
  interactions;

 Generally very reactive chemicals, often with relatively
  short environmental half-lives;

 Tend to be highly reactive electrophiles;

 Typically act at the site of first contact;
Direct-acting mutagens

 Chemical stability, transport, membrane permeability
  generally determine the degree of mutagenicity
  associated with these agents;

 Direct-acting mutagens are generally mutagenic at
  multiple tissue sites and in essentially all species;
Direct-acting mutagens

 Classical examples:
   Nitrogen and sulfur mustards;
   Methyl methane sulfonate;
   Propane sulfone,
   Ethyleneimine
   Beta propiolactone
   Dimethylsulfate
Classical Structural Alerts for Reactive, Electrophilic Direct-Acting
                               Mutagens




                                                       Cl-CH2-O-CH2-Cl

                                                       Haloethers

Epoxides         Strained lactones      Sulfonates




                              Enals
Mechanisms of Chemical Interaction
with DNA: Electrophiles

 The majority of genotoxic chemicals that
  directluy interact with DNA are either
  electrophiles or are metabolized to
  electrophiles;
 Electrophiles are positively charged species
  that are attracted to an electron rich
  center i.e. a nucleophile;
Mechanisms of Chemical Interaction
with DNA: Electrophiles

 An electrophile (literally electron-lover) is a
  reagent attracted to electrons that
  participates in a chemical reaction by
  accepting an electron pair in order to bond
  to a nucleophile;
 Most electrophiles are positively charged,
  have an atom that carries a partial positive
  charge, or have an atom that does not
  have an octet of electrons;
Mechanisms of Chemical Interaction
with DNA: Electrophiles

 Specific areas of DNA behaves like a nucleophile;
 DNA nucleophilic centers:
   Ring nitrogens of the DNA bases are generally the
    most reactive centers;
   Ring oxygens of the DNA bases are also reactive
    centers;
   S: groups in DNA are also targets
   Critically, the N7 of guanine (most reactive); the N3 of
    adenine and the O6 of guanine are the most common
    sites
Nucleophilic Centers of DNA Bases
Mechanisms of Chemical Interaction
with DNA: Electrophiles
 Electrophilic damage may also occur to the
  phosphodiester backbone of DNA:
Mechanisms of Chemical Interaction
with DNA: Electrophiles
 Different electrophiles display different
  preferences for the various DNA nucleophilic
  sites and different spectra of damage;


 One of the intensively researched concepts is
  that the DNA damage spectrum for different
  electrophiles could be used as a marker of
  environmental exposure: has not worked
  because of the variable ability of DNA to repair
  different types of DNA damage plus the often
  tissue-specific nature of mutation spectra.
Mechanisms of Chemical Interaction
with DNA: Oxidization
 Oxidation of DNA bases is a normal
  background biological event associated with
  aerobic metabolism and other cell reactions;


   Important endogenous oxidizing agents are:
    H2O2, superoxide anion, nitric oxide species, lipid
    peroxides, hydroxyl radicals, Fenton reaction
    products;
Mechanisms of Chemical Interaction
with DNA: Oxidization
 Endogenous oxidative damage to DNA occurs
  at a rate of about 120 occurrences per cell per
  hour in NORMAL cells!
 In general, oxidative damage to DNA is
  repaired with high fidelity at a maximal rate of
  about 105 base pairs per hour;
 Critically, there are a number of antioxidant
  pathways that limit the amount of
  endogenously oxidative species that are
  produced under normal circumstances;
Mechanisms of Chemical Interaction
with DNA: Oxidization
 Oxidative damage to DNA occurs most readily
  at guanine residues due to the high oxidation
  potential of this base relative to cytosine,
  thymine, and adenine;

 Most common oxidized DNA adduct is 8-
  hydroxyguanine followed by thymine glycol.
Mechanisms of Chemical Interaction
with DNA: Hydrolysis - deamination,
depurination, and depyrimidination
 Deamination of cytosine:
   Spontaneous deamination is the hydrolysis
    reaction of cytosine into uracil,
   Corrected for by the removal of by uracil-DNA
    glycosylase, generating an abasic (AP) site;
   The AP site is then corrected by base excision
    repair;
Mechanisms of Chemical Interaction
with DNA: Hydrolysis -
deamination, depurination, and
depyrimidination
 5-methylcytosine: Spontaneous deamination of
  5-methylcytosine results in thymine;
   This is the most common single nucleotide
    mutation. In DNA, this reaction can be corrected
    by the enzyme thymine-DNA glycosylase;
Mechanisms of Chemical Interaction
with DNA: Hydrolysis - deamination,
depurination, and depyrimidination
 Guanine: Deamination of guanine results in the
  formation of xanthine.
   Xanthine, in a manner analogous to the enol
    tautomer of guanine, selectively base pairs with
    thymine instead of cytosine. This results in a post-
    replicative transition mutation, where the original
    G-C base pair transforms into an A-T base pair.
    Correction of this mutation involves the use of
    alkyladenine glycosylase during base excision
    repair;
Mechanisms of Chemical Interaction
with DNA: Hydrolysis - deamination,
depurination, and depyrimidination
 Adenine: Deamination of adenine results in the
  formation of hypoxanthine.
   Hypoxanthine, in a manner analogous to the
    imine tautomer of adenine, selectively base pairs
    with cytosine instead of thymine. This results in a
    post-replicative transition mutation, where the
    original A-T base pair transforms into a G-C base
    pair;
Mechanisms of Chemical Interaction
with DNA: Hydrolysis -
deamination, depurination, and
depyrimidination
 Depurination is a chemical reaction of purine
  deoxyribonucleosides, deoxyadenosine and
  deoxyguanosine, in which the β-N-glycosidic bond is
  hydrolytically cleaved releasing a nucleic base,
  adenine or guanine, respectively.
 Deoxyribonucleosides and their derivatives are
  substantially more prone to depurination than their
  corresponding ribonucleoside counterparts.
 Loss of pyrimidine bases (Cytosine and Thymine)
  occurs by a similar mechanism, but at a
  substantially lower rate.
Mechanisms of Chemical Interaction
with DNA: Hydrolysis - deamination,
depurination, and depyrimidination
 When depurination occurs with DNA, it leads to the
  formation of apurinic site and results in an alteration
  of the structure;

 As many as 5,000 purines are lost this way each day
  in a typical human cell;

 [In cells, one of the main causes of depurination is
  the presence of endogenous metabolites
  undergoing chemical reactions;
Mechanisms of Chemical Interaction
with DNA: Hydrolysis - deamination,
depurination, and depyrimidination
 Apurinic sites in double-stranded DNA are efficiently
  repaired by portions of the base excision repair
  (BER) pathway;
 Depurinated bases in single-stranded DNA
  undergoing replication can lead to mutations,
  because in the absence of information from the
  complementary strand, BER can add an incorrect
  base at the apurinic site, resulting in either a
  transition or transversion mutation;
 Depurination is known to play a major role in cancer
  initiation.
Mechanisms of Chemical Interaction
with DNA: Intercalation
 Intercalation occurs when ligands of an
  appropriate size and chemical nature fit
  themselves in between base pairs of DNA;

 Intercalating agents are generally
  polycyclic, aromatic, and planar, and good
  DNA stains;

 Important examples:
   Ethidium bromide (DNA stain);
   Anticancer agents:
    proflavine, daunorubicin, doxorubicin, dactinomy
    cin, thalidomide.
Molecular structure of ethidium intercalated between two pairs of adenine-
                              uracil base pairs.
Mechanisms of Chemical Interaction
with DNA: Intercalation
 In order for an intercalator to fit between base pairs,
  the DNA must dynamically open a space between
  its base pairs by unwinding;
 The amount of unwinding depends on the specific
  agent;
 This unwinding induces local structural changes to
  the DNA strand resulting in functional changes:
  inhibition of transcription and replication and DNA
  repair processes,
 Intercalating agents are commonly potent
  mutagen.
Mechanisms of Chemical Interaction
with DNA: Intercalating agents
 Important examples:
   8,9 epoxide of aflatoxin B1;
   Acridine dyes.
Mechanisms of Chemical Interaction
with DNA: DNA cross linking
 Crosslinks occur when exogenous or endogenous
  agents react with two different positions in the DNA;

 Crosslinks can occur in the same DNA strand
  (intrastrand crosslink) or between opposite strands
  (interstrand crosslink);

 Crosslinks between DNA and protein can occur;
Mechanisms of Chemical Interaction
with DNA: DNA cross linking
 Crosslinks impair DNA replication if the crosslink is not
  repaired;

 Mechanisms:
   Bifunctional alkylating agents (e.g. methylene
    dimethanesulphonate, sulphur mustard, methyl
    methanesulphonate): mostly act on adjacent N7-
    guanine bases;
   Cisplatin: 1,2-intrastrand d(GpG) adducts (via N7-
    guanine bases);
Mechanisms of Chemical Interaction
with DNA: DNA cross linking
 Mechanisms:
   Nitrous acid is formed in the stomach from
    dietary nitrites (meat prreservatives): forms
    interstrand DNA crosslinks at the aminogroup
    of N2 of guanine at CG sequences;

   Malondialdehyde from lipid peroxidation:
    forms etheno adduct-derived interstrand
    crosslinks;
Mechanisms of Chemical Interaction
with DNA: DNA cross linking
 Mechanisms:
   Psoralens: photoactivated by UVA  form
    covalent adducts with thymine, one type of
    which is an intrastrand crosslinking reaction
    targets TA sequences intercalating in DNA
    and linking one base of the DNA with the one
    below it. Psoralen adducts cause replication
    arrest and is used in the treatment of psoriasis
    and vitiligo;
Mechanisms of Chemical Interaction
with DNA: DNA cross linking
 Mechanisms:
   Aldehydes such as acrolein and
    crotonaldehyde found in tobacco smoke or
    automotive exhaust can form DNA
    interstrand crosslinks in DNA.

   Formaldehyde (HCHO) induces protein-DNA
    and protein-protein crosslinks;
Mechanisms of Chemical Interaction
with DNA: DNA single strand breaks
 Single strand breaks are an extremely common
  phenomenon – thousands of incidents per cell
  per day;

 Mechanisms of production:
   Free radical attack (notably with radiation-
    induced DNA damage);
   DNA alykylation (i.e. electrophylic attack);
   Many of the mechanisms are poorly
    understood;
Mechanisms of Chemical Interaction
with DNA: DNA double strand breaks
 Particularly hazardous to the cell because they can
  lead to genome rearrangements: regarded as the
  most dangerous of DNA lesions;
 Mechanisms are poorly understood, however
  oxidization and alkylating agents are able to
  produce these DNA lesions;
 DSBs are induced by a number of different
  mechanisms, including exposure to ionizing
  radiation, radiomimetic drugs, collapse of
  replication forks when the replication machinery
  encounters single-stranded breaks (SSBs) in the
  template DNA,
Micro-DNA Lesions:
     Small damage with BIG outcomes.
The Ability to Chemically Interact
With DNA is Not Enough
 For a mutation to occur:
   Exposure of DNA generally must occur at the right
    time in the cell cycle;

   A DNA lesion must be chemically produced;

   The DNA lesion must not be so gross as to prevent
    DNA replication and/or produce cell death;
The Ability to Chemically Interact
With DNA is Not Enough
 For a mutation to occur:
   The lesion must persist in the DNA long enough for at
    least 1 cell division (i.e. the “fixing” of the mutation
    within the cell genome; often referred to as the
    “expression time” in genetox assays);

   The DNA lesion must not trigger the G1/S checkpoint
    (i.e. apoptosis or senescence; more on this in the
    carcinogenesis sildes);

   The DNA lesion must not trigger the intra-S phase
    checkpoint (if it does, repair is the likely outcome);
The Cell Cycle and Mutation:
    The vulnerability of the S phase.
 Small mutations are most likely to occur when
  the DNA is being copied i.e. S phase;

 Reason for this is that the DNA double helix is
  unwound and the 2 strands are separated 
  single DNA strands are particularly vulnerable to
  chemical attack;
(A) Nucleoside triphosphates serve as a substrate for DNA polymerase, according to the
mechanism shown on the top strand. Each nucleoside triphosphate is made up of three
phosphates (represented here by yellow spheres), a deoxyribose sugar (beige rectangle)
and one of four bases (differently colored cylinders). The three phosphates are joined to
each other by high-energy bonds, and the cleavage of these bonds during the
polymerization reaction releases the free energy needed to drive the incorporation of
each nucleotide into the growing DNA chain. The reaction shown on the bottom strand,
which would cause DNA chain growth in the 3' to 5' chemical direction, does not occur in
nature. (B) DNA polymerases catalyse chain growth only in the 5' to 3' chemical direction,
but both new daughter strands grow at the fork. The leading strand grows continuously,
whereas the lagging strand is synthesized by a DNA polymerase through the backstitching
mechanism illustrated. Thus, both strands are produced by DNA synthesis in the 5' to 3'
direction.
Proteins at the Y-shaped DNA replication fork: These proteins are illustrated schematically in panel a of the figure below, but in reality, the
fork is folded in three dimensions, producing a structure resembling that of the diagram in the inset b. Focusing on the schematic illustration
in a, two DNA polymerase molecules are active at the fork at any one time. One moves continuously to produce the new daughter DNA
molecule on the leading strand, whereas the other produces a long series of short Okazaki DNA fragments on the lagging strand. Both
polymerases are anchored to their template by polymerase accessory proteins, in the form of a sliding clamp and a clamp loader. A DNA
helicase, powered by ATP hydrolysis, propels itself rapidly along one of the template DNA strands (here the lagging strand), forcing open
the DNA helix ahead of the replication fork. The helicase exposes the bases of the DNA helix for the leading-strand polymerase to copy.
DNA topoisomerase enzymes facilitate DNA helix unwinding. In addition to the template, DNA polymerases need a pre-existing DNA or
RNA chain end (a primer) onto which to add each nucleotide. For this reason, the lagging strand polymerase requires the action of a DNA
primase enzyme before it can start each Okazaki fragment. The primase produces a very short RNA molecule (an RNA primer) at the 58
end of each Okazaki fragment onto which the DNA polymerase adds nucleotides. Finally, the single-stranded regions of DNA at the fork
are covered by multiple copies of a single-strand DNA-binding protein, which hold the DNA template strands open with their bases
exposed. In the folded fork structure shown in the inset, the lagging-strand DNA polymerase remains tied to the leading-strand DNA
polymerase. This allows the lagging-strand polymerase to remain at the fork after it finishes the synthesis of each Okazaki fragment. As a
result, this polymerase can be used over and over again to synthesize the large number of Okazaki fragments that are needed to produce
a new DNA chain on the lagging strand. In addition to the above group of core proteins, other proteins (not shown) are needed for DNA
replication. These include a set of initiator proteins to begin each new replication fork at a replication origin, an RNAseH enzyme to remove
the RNA primers from the Okazaki fragments, and a DNA ligase to seal the adjacent Okazaki fragments together to form a continuous DNA
strand.
The Cell Cycle and Mutation:
    The S phase checkpoint.
 The S-phase checkpoint is a surveillance
  mechanism, mediated by the protein kinases ATR
  and Chk2 in human cells;
 Responds to to DNA damage by co-ordinating a
  global cellular response necessary to maintain
  genome integrity;
 A key aspect of this response is the stabilization of
  DNA replication forks, which is critical for cell
  survival;
 A defective checkpoint causes irreversible
  replication-fork collapse and leads to genomic
  instability, a hallmark of cancer cells.
If DNA replication is blocked (e.g. by a DNA adduct) ssDNA regions at stalled
forks continue to grow because MCM (minichromosome maintenance complex)
helicase continues DNA unwinding, although uncoupled from DNA synthesis.

The ssDNA binds RPA (replication protein A), which triggers the activation of the
checkpoint response. This process is initiated by the recruitment of the Mec1/ATR
sensor to RPA-coated ssDNA at stalled forks by its regulatory subunit, Ddc2 (ATRIP
in human cells). Mec1 then phosphorylates Mrc1 (the homologue of human
Claspin), a mediator that transduces the signal from Mec1 to the effector kinase
Rad53, which becomes phosphorylated and activated.
The Cell Cycle and Mutation:
    The S phase checkpoints.
 The S-phase checkpoint response co-ordinates DNA
  replication, DNA repair and cell-cycle progression and
  regulates processes such as firing of replication origins,
  stabilization of DNA replication forks in response to DNA
  damage or replicative stress, resumption of stalled DNA
  replication forks, transcriptional induction of DNA
  damage response genes, choice of the repair pathway
  and inhibition of mitosis until replication is completed;



 The S-phase checkpoint is required for cellular viability in
  DNA damage or replicative-stress conditions
Types of Mutations: Point mutations.

 Point mutation = single base substitution = the
  replacement of a single base nucleotide with another
  nucleotide of the genetic material (either DNA or RNA);

 Point mutations most commonly occur during S phase
  (i.e. DNA replication);

 The term also includes insertions or deletions of a single
  base pair which will result in a frame shift mutation;

 Classifications:
   By type: deletion, transition, insertion, or transversion;
   By the effect on function: nonsense, missense and silent;
Types of Mutations: Point mutations.

 Transversions (beta mutations):
   The substitution of a purine for a pyrimidine or vice versa;
   Can only be repaired by a spontaneous reversion;
   Transitions produce large chemical changes to DNA
    structure; thus the consequences of this change tend to be
    more drastic than those of transitions.
   Transversions are classically caused by ionizing radiation and
    alkylating agents.
Types of Mutations: Point mutations.
 Transitions (alpha mutations):
   A point mutation that changes a purine nucleotide to another
    purine (A ↔ G) or a pyrimidine nucleotide to another pyrimidine (C
    ↔ T);
   Approximately two out of three single nucleotide polymorphisms
    (SNPs) are transitions;
   Transitions can be caused by oxidative deamination and
    tautomerization;
   Although there are twice as many possible transversions, transitions
    appear more often in genomes, possibly due to the molecular
    mechanisms that generate them;
   5-Methylcytosine is more prone to transition than unmethylated
    cytosine, due to spontaneous deamination.
Types of Mutations: Point mutations.

 Nonsense mutations are point mutations in a sequence of DNA
  that results in a premature stop codon, or a nonsense codon in
  the transcribed mRNA, and in a truncated, incomplete, and
  usually nonfunctional protein product;
Types of Mutations: Point mutations.
 Missense mutations are point mutations in which a single
  nucleotide is changed, resulting in a codon that codes for a
  different amino acid i.e. a non-synonymous change (mutations
  that change an amino acid to a stop codon are considered
  nonsense mutations, rather than missense mutations). There are
  2 possible outcomes:
   Conservative mutations: Result in an amino acid change.
    However, the properties of the amino acid remain the same
    (e.g., hydrophobic, hydrophilic, etc). At times, a change to one
    amino acid in the protein is not detrimental to the organism as a
    whole. Most proteins can withstand one or two point mutations
    before their functioning changes;
   Non-conservative mutations: Result in an amino acid change that
    has different properties than the wild type. The protein may lose its
    function, which can result in a disease in the organism.
Types of Mutations:
        Frame shift mutations.
 Silent mutations: Code for the same amino acid.
   A silent mutation has no effect on the functioning of the protein;
   A single nucleotide can change, but the new codon specifies
    the same amino acid, resulting in an non-mutated protein;
   This type of change is also called synonymous change, since the
    old and new codon code for the same amino acid;
   This is possible because 64 codons specify only 20 amino acids;
   Different codons can lead to differential protein expression
    levels.
Types of Mutations: Point mutations.
 A frame shift mutation (also called a framing error or a reading
  frame shift) is a genetic mutation caused by indels (insertions or
  deletions) of a number of nucleotides that is not evenly divisible by
  three from a DNA sequence;

 Remember that it takes 3 DNA and RNA nucleotides to code for a
  specific amino acid in a protein i.e. the “reading frames” consist of
  3 nucleic acid residues;

 Thus insertion or deletion of a DNA nucleotide can change the
  reading frame (i.e. the grouping “3s” of the codons), resulting in a
  completely different translation from the original;The earlier in the
  sequence the deletion or insertion occurs, the more altered the
  protein produced is.
Types of Mutations: Point mutations.
 Frame shift mutations;


   The earlier in the sequence the deletion or insertion
    occurs, the greater the effect on protein structure. This is
    because all of the reading frames after the insertion or
    deletion will be altered i.e. earlier in the relevant DNA coding
    sequence the error is, the bigger the overall change to the
    downstream amino acid composition of the protein;
Types of Mutations: Point mutations.
 Frame shift mutations;
   Frameshift mutations will also alter the first stop codon ("UAA",
    "UGA" or "UAG") encountered in the sequence. The
    polypeptide being created could be abnormally short or
    abnormally long, and will most likely not be functional;

   Frameshift mutations frequently result in severe genetic
    diseases
DNA Repair Mechanisms.
 High level overview only;

 The self-repair of DNA is unique amongst biological molecules;

 Major types of mammalian DNA repair:
   Base excision repair;
   Nucleotide excision repair;
   Mismatch repair;
   Recombinational repair.
DNA Repair Mechanisms.
 DNA repair capacity varies by mechanism, tissue, organ,
  individual and species;

 Not all DNA adduct types are equal: some are more easily
  repaired than others and some types cannot be repaired;

 Because of the above 2 points, the use of the number of DNA
  adducts per cell is NOT a reliable predictor of genetic hazard
  UNLESS there is very detailed information regarding the kinetics
  of DNA adduct removal in the specific target tissue and target
  species!
DNA Repair Mechanisms.

 In general terms, DNA repair are saturatable mechanisms i.e.
  there is maximal threshold for the amount of DNA that can be
  repaired within a given set of parameters;

 All DNA repair mechanisms have the ability to detect and repair
  DNA, and if the DNA repair is successful, the impact of the
  original DNA damage on the animal is reduced or eliminated;
DNA Repair Mechanisms:
 In general terms, when low levels of DNA damage occur (i.e.
  below saturation for repair), error-free (high fidelity) repair
  occurs. When there are large amounts of DNA damage (i.e.
  above the saturation for repair), error prone DNA repair
  predominates;


   However: there are big species, sex, age, tissue, and organ
    differences;
   The type of DNA adduct has a significant influence over
    which type of repair predominates;
DNA Repair Mechanisms:
 Intrinsic variability within human populations
   There is a very large variability to repair DNA
     damage between individuals: up to 65% of
     average rate in populations without inherited DNA
     repair defects (e.g. XP);
      People with XP have DNA repair capacity of ~1-
        2% of normal;
   There are differences in DNA repair capacity
     between rodents and humans: you cannot directly
     extrapolate results unless you accurate kinetics
     across the relevant species;
DNA Repair Mechanisms:
   Mechanisms where only one strand is
   damaged
 When only one of the two strands of a double helix
  has a defect (i.e. only one of the 2 strands has a
  damaged/missing nucleotide), the other strand can
  be used as a template to guide the correction of the
  damaged strand;

 These types of DNA repair are called excision repair:
  remove the damaged nucleotide and replace it with
  an undamaged nucleotide complementary to that
  found in the undamaged DNA strand
DNA Repair Mechanisms:
       Base excision repair
 Base excision repair:
   Repairs damage to a single base caused by oxidation,
    alkylation, hydrolysis, or deamination;
   The damaged base is removed by a DNA glycosylase;
   The DNA base is then recognized by an enzyme called
    AP endonuclease, which cuts the phosphodiester
    bond;
   The missing part is then resynthesized by a DNA
    polymerase, and a DNA ligase performs the final nick-
    sealing step;
Base Excision Repair
BER: Short Patch
BER: Long Patch
DNA Repair Mechanisms:
       Base excision repair
 Base excision repair:
   BER is the major repair pathway involved in the removal
    of non-bulky damaged nucleotides;
   In general BER is a high fidelity process i.e. not error
    prone;
   BER protects both nuclear and mitochondrial DNA;
   Heritable defects in BER (particularly DNA polymerase)
    are associated with cancer.
DNA Repair Mechanisms:
      Nucleotide excision repair
 Nucleotide excision repair (NER), which recognizes
  bulky, helix-distorting lesions;

 A specialized form of NER known as transcription-
  coupled repair deploys NER enzymes to genes that
  are being actively transcribed;
1) 3 protein complexes
are involved in DNA-
damage recognition:
XPA, XPC-HR23 and RPA.
2) These proteins recruits
Transcription factor II
(TFIIH) that incorporate
two helicases: XPB and
XPD that unwinds a 30 bp
DNA fragment around
the DNA damage.
3) After DNA unwinding,
damaged-DNA strand is
excised by XPG and the
XPF-ERCC1 complex at 3'
and 5' sites respectively.

4) After excision,
damaged-DNA strand is
removed and replaced
by re-synthesizing the
template complementary
DNA strand by
polymerase complex (Pol.
E/D, replication protein A
(RPA) and replication
factor C).
DNA Repair Mechanisms:
      Nucleotide excision repair
 Mismatch repair (MMR), which corrects errors of DNA
  replication and recombination that result in
  mispaired (but undamaged) nucleotides;

 MMR functions primarily as a “proof reader” following
  DNA replication
Mismatch Repair
DNA Repair Mechanisms:
      Nucleotide excision repair
 Mismatch repair (MMR), which corrects errors of DNA
  replication and recombination that result in
  mispaired (but undamaged) nucleotides;

 MMR functions primarily as a “proof reader” following
  DNA replication
Macro DNA Damage:
 Macro DNA damage = damage to chromosomes =
  clastenogenesis e.g. single strand breaks, double
  strand breaks, sister chromatid exchange, non-
  homologous end joining, changes in ploidy;
   Abnormal chromosome number = aneuploidy;
   Increased chromosome number = polyploidy;
Macro DNA Damage:
       Sister chromatid exchange
 Exchange of genetic material between two identical
  sister chromatids or between chromosomes with identical
  mutations;

 Primarily occurs during S phase;

 Four to five sister chromatid exchange/chromosome
  pair/mitosis is within the normal range, 14-100 exchanges
  is not normal and presents a danger to the organis;

 Mediated by the homologous end-joining mechanism fo
  DNA;
Macro DNA Damage:
       Sister chromatid exchange
       (equal cross over)
 SCEs can be induced by various genotoxic treatments
  that result in double DNA strand breaks, suggesting that
  SCEs reflect a DNA repair process i.e. they are measure of
  DNA damage;

 This process is considered to be conservative and error-
  free, since no information is generally altered during
  reciprocal interchange by homologous recombination.

 Most forms of DNA damage induce chromatid exchange
  upon replication fork collapse: Holliday model;

 Occurs during prophase I of meiosis (pachytene) in a
  process called synapsis.
Macro DNA Damage:
      Other potential outcomes of
      DNA double strand breaks
 SCE (equal cross over) is the least harmful outcome
  of a DSB;

 Other potentially catestrophic outcomes include due
  to misrepair of DSBs include:
   Inversions;
   Interstitial deletions;
   Terminal deletions;
   Translocations;
   Unequal crossovers.
Macro DNA Damage:
         Other potential outcomes of
         DNA double strand breaks
 Inversions:
   Inversions that
    involve the
    centromere are
    called pericentric
    inversions;
   Those that do not
    involve the
    centromere are
    called paracentric
    inversions;
   Inversions potentially
    have massive effects
    on gene function.
Macro DNA Damage:
          Other potential outcomes of
          DNA double strand breaks
 Interstitial deletions:
   Causes include the
    following: Losses
    from translocation;
    chromosomal
    crossovers within a
    chromosomal
    inversion; unequal
    crossing over;
    breaking without
    rejoining
Macro DNA Damage:
    Other potential outcomes of
    DNA double strand breaks
Macro DNA Damage:
         Other potential outcomes of
         DNA double strand breaks
 Translocation
   = a chromosome abnormality caused by rearrangement of
     parts between nonhomologous chromosomes;

   Gene fusion may be created when the translocation joins
    two otherwise separated genes: very important in
    carcinogenesis as it may place the coding region of a
    relatively inactive gene with normally very active gene
    promoter region  inappropriate upregulation of a gene
    (alternatively gene silencing may occur);
Macro DNA Damage:
          Other potential outcomes of
          DNA double strand breaks
 Translocation:
   There are two main types, reciprocal (also known as non-
     Robertsonian) and non-reciprocal (Robertsonian);

   Also, translocations can be balanced (in an even exchange
    of material with no genetic information extra or missing, and
    ideally full functionality) or unbalanced (where the exchange
    of chromosome material is unequal resulting in extra or
    missing genes).
Macro DNA Damage: Other potential outcomes of
      DNA double strand breaks




 Reciprocal translocations:
   Reciprocal translocations are usually an exchange of
    material between nonhomologous chromosomes;
   Estimates of incidence range from about 1 in 500 human
    newborns;
   Such translocations are usually harmless and may be found
    through prenatal diagnosis;
   However, carriers of balanced reciprocal translocations have
    increased risks of creating gametes with unbalanced
    chromosome translocations leading to miscarriages or
    children with abnormalities.
   Most balanced translocation carriers are healthy and do not
    have any symptoms.
Macro DNA Damage: Other potential
    outcomes of DNA double strand breaks




 Nonreciprocal (Robertsonian) translocations:
   This type of rearrangement involves two acrocentric
    chromosomes that fuse near the centromere region with loss
    of the short arms
   The resulting karyotype in humans leaves only 45
    chromosomes since two chromosomes have fused together
Macro DNA Damage: Other potential
    outcomes of DNA double strand breaks




 Nonreciprocal (Robertsonian) translocations:
   This has no direct effect on the phenotype since the only
    genes on the short arms of acrocentrics are common to all of
    them and are present in variable copy number (nucleolar
    organiser genes). Robertsonian translocations have been
    seen involving all combinations of acrocentric chromosomes.
    The most common translocation in humans involves
    chromosomes 13 and 14 and is seen in about 0.97 / 1000
    newborns.
   Carriers of Robertsonian translocations are not associated
    with any phenotypic abnormalities, but there is a risk of
    unbalanced gametes which lead to miscarriages or
    abnormal offspring.
Macro DNA Damage: Other potential
     outcomes of DNA double strand breaks




 Balanced translocations: no genetic material is lost
Macro DNA Damage: Other potential
     outcomes of DNA double strand breaks




 Unbalanced translocation: genetic material is lost from one
  chromosome but gained by another. This means that the
  progeny will either have missing genetic material or extra
  genetic material
Macro DNA Damage:
       Other potential outcomes of
       DNA double strand breaks




 Acentric fragments: a segment of a chromosome
  that lacks a centromere;
Macro DNA Damage:
          Other potential outcomes of
          DNA double strand breaks

 Acentric fragments:
   Because acentric fragments lack a centromere, the cannot attach
    to the mitotic spindle i.e. acentric fragments are not evenly
    distributed to the daughter cells in cell division (mitosis and meiosis).
    As a result one of the daughters will lack the acentric fragment;
   Lack of the acentric fragment in one of the daughter cells
    may have deleterious consequences, depending on the
    function of the DNA in this region of the chromosome;
   In the case of a gamete, it will be fatal if essential DNA is
    contained in that DNA segment;
   In the case of a diploid cell, the daughter cell lacking the
    acetric fragment will show expression of any recessive genes
    found in the homologous chromosome.
Acentric fragments are lost from the nuclei of cells following
mitosis or meiosis. They form a micronucleus.
Macro DNA Damage: Changes in
     chromosome number

 Changes to chromosome number can result from:
   Nonreciprocal (Robertsonian) translocations;
   Errors in chromosomal segregation i.e. a whole
    chromosome is left behind during anaphase of
    mitosis or meiosis;
   Damage to the mitotic spindle: e.g. griseofulvin,
    pcalitaxel, colecemid, vinblastine;
Macro DNA Damage: Changes in
      chromosome number

 Kinetocore:
   The protein structure on chromatids where the
     spindle fibers attach during cell division to pull sister
     chromatids apart;
   The kinetochore forms in eukaryotes, assembles on
     the centromere and links the chromosome to
     microtubule polymers from the mitotic spindle
     during mitosis and meiosis;
   Kinetochores start, control and supervise the
     striking movements of chromosomes during cell
     division;
Macro DNA Damage: Changes in
      chromosome number

 Kinetocore:
   The protein structure on chromatids where the
     spindle fibers attach during cell division to pull sister
     chromatids apart;
   The kinetochore forms in eukaryotes, assembles on
     the centromere and links the chromosome to
     microtubule polymers from the mitotic spindle
     during mitosis and meiosis;
   Kinetochores start, control and supervise the
     striking movements of chromosomes during cell
     division;
Macro DNA Damage: Changes in
     chromosome number

 Kinetocore:

   Kinetochores are critical in initiating/avoiding the
    spindle checkpoint
Macro DNA Damage: Changes in
      chromosome number

 The spindle checkpoint:
   The spindle checkpoint (= spindle assembly checkpoint = mitotic
    checkpoint), is a cellular mechanism responsible for detection of:
      Correct assembly of the mitotic spindle
      Attachment of all chromosomes to the mitotic spindle in a
       bipolar manner
      Congression of all chromosomes at the metaphase plate.
   When just one chromosome (for any reason) remains lagging during
    congression, the spindle checkpoint machinery generates a delay
    in cell cycle progression: the cell is arrested, allowing time for repair
    mechanisms to solve the detected problem. After some time, if the
    problem has not been solved, the cell will be targeted for apoptosis
    (programmed cell death), a safety mechanism to avoid the
    generation of aneuploidy, a situation which generally has dramatic
    consequences for the organism.
Macro DNA Damage: Changes in
     chromosome number

 Micronuclei can also contain whole chromosomes that
  were improperly attached to the mitotic spindle during
  anaphase: these are detectable by staining the
  micronuclei for kinetocores;

 There are at least major causes of micronuclei:
   The formation of chromosome fragments that lack a
    centromere (and thus a kinetocore  cannot attach o the
    mitotic spindle);
   The loss of a whole chromosome which has failed to attach
    to the mitotic spindle or has broken off the mitotic spindle.
    This effect is produced by spindle agents;
   Extrachromosomal double minutes.
Macro DNA Damage: Changes in
      chromosome number

 So what the heck is a double minute you ask (no it is not 120
  seconds):
 Double minutes are small fragments of extrachromosomal DNA,
  which have been observed in a large number of human tumors;
 They are a manifestation of gene amplification during the
  development of tumors, which give the cells selective advantages
  for growth and survival;
 They frequently harbor amplified oncogenes and genes involved in
  drug resistance;
 Double minutes, like actual chromosomes, are composed of
  chromatin and replicate in the nucleus of the cell during cell
  division;
 Unlike typical chromosomes, they are composed of circular
  fragments of DNA, up to only a few million base pairs in size and
  contain no centromere or telomere.
Micronucleus
fluorescently labeled
for kinetocores
Mechanisms of micronuclei formation. (A)
Aneugenic agents prevent the formation of
the spindle apparatus during mitosis. The use of
these agents generates micronuclei as a
consequence of whole chromosomes lagging
behind at anaphase. These chromosomes are
left out of the cell nucleus at the end of mitosis;
The DNA in the micronuclei could balance the
nuclear DNA and result in a complete
genome, or be additional to the cell's
genome.
 (B) Clastogenic agents induce micronuclei by
breaking the double helix of DNA, thereby
forming acentric fragments. These fragments
are incapable of adhering to the spindle fibres
and integrating in the daughter nuclei, and
are thus left behind during mitosis.
(C) Micronuclei can also contain highly
amplified gene sequences, derived from
extrachromosomal double minutes (DM)
(yellow dots indicate the presence of a DM).
(D(I)) Torsion between the two centromeres of
a dicentric chromosome would give rise to the
formation of an anaphase bridge that is
frequently resolved by breakage. The bridge
breakage often results in the formation of
acentric fragments that are not included in
any of the daughter cell nuclei and form one
or more micronuclei at the end of mitosis. (D
(II)) It has also been described that, instead of
breaking, dicentric chromosomes involved in
anaphase bridges are sometimes detached
from the two centrosomes, left behind at
anaphase and sequestered into micronuclei.
US EPA Testing Requirements & Tiers
Tier   Test Types                                     Assessment Function

                                                      Rapid, low cost screening,
       Ames (bacterial reverse mutation)
                                                      typically for agents where
       In vitro mammalian cell mutation (e.g.
                                                      human exposure is low.




                                                                                     Hazard Identification
 1     mouse lymphoma TK)
                                                      If all results are negative,
       In vitro chromosome aberration or
                                                      generally no further
       micronucleus assay
                                                      testing is required

       In vivo testing: at least one or more of: in   Required if there is
       vivo micronucleus, comet assay, in vivo        significant human
 2     DNA binding, in vivo unscheduled DNA           exposure or +ve results in
       synthesis, transgenic mouse models             Tier 1
                                                      Required if +ve results in
       In vivo tests in germ cells (i.e. dominant
                                                      Tier 2. Provides a basis for
 3     lethal, germ cell micronucleus, germ cell
                                                      hazard assessment of
       DNA binding, germ cell USD,
                                                      germ line effects
                                                      Provides a quantitative
       Quantitative in vivo tests for germ cell
                                                      assessment of germ cell
       mutation (specific locus test, visible or
 4     biochemical markers [mouse spot],
                                                      mutations for use in
                                                      quantitative risk
       heritable translocation test in mice)
                                                      assessments
Germ Cell Versus Somatic Mutation:
        Female germ cell
 The timing of exposure is critically important:


   Mutation is most likely to occur during the S phase
    (during DNA replication) i.e. mitosis and meiosis;
Germ Cell Versus Somatic Mutation:
       Female germ cell
 The timing of exposure is critically important:
   In humans (and most mammals), this means:
     During mitosis of oogonium during fetal
       development (between weeks 4 and 30 in
       humans; between days 14.5 and 18.5 in the rat
       and between days 10.5-12.5 in mice);
     There are no further S phases until after
       fertilization and the formation of a zygote;

   Remember: DNA repair occurs in oocytes!
Germ Cell Versus Somatic Mutation:
   Female germ cell
Germ Cell Versus Somatic Mutation:
       Female germ cell
 Consequences in terms of germ cell mutation:
   The critical timing for female germ cell mutation in
    mammals occurs at the prenatal stage of
    development!

   Ooctyes are RESISTANT to mutation by non-
    radiomimetic chemicals (i.e. chemicals that do not
    produce chromosome or chromatid breaks);

   Oocytes are susceptible to radiation and
    radiomimetic chemicals;
Germ Cell Versus Somatic Mutation:
       Male germ cells
 Same basic principle holds true: S phase of cell
  division processes is most susceptible to
  mutagenesis;
 S phase in spermatogenesis occurs during
  spermatogonial stem cell stage of development;
 Unlike in females, spermatogonial stem cell
  mitosis occurs throughout the life time of males!
 Late spermatids and spermatozoa lack DNA
  repair – also susceptible to unrepaired DNA
  damage!
Germ Cell Versus Somatic Mutation
 Overall, males are generally regarded as being
  at greater risk of generating germ cell mutations
  than females because of the continuous, life-
  time replication of spermatogonial stem cells
  (where as oogonium are only generated in large
  numbers during prenatal development in
  females).
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Fundamental principles:
   These assays are for micro-DNA damage;

   Mutant bacterial test strains are created/selected (typically
    loose the capacity for synthesis of an amino acid) so they
    require some form of additional supplementation (usually an
    amino acid) in order to grow – these are called auxotrophs
    (auxotrophy is the inability of an organism to synthesize a
    particular organic compound required for its growth);
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Fundamental principles:

   The mutation required to produce the auxotrophic strain is
    generally either a point mutation or a frame shift mutation i.e.
    a small DNA sequence change.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Fundamental principles:
   In order for an auxotroph to grow in medium where the
    supplement is NOT present, they must undergo a reverse
    mutation in order to regain the capacity to synthesize the
    essential substance for growth;

   Once the bacteria have undergone a reverse mutaiton, they
    are able to grow in media that DO NOT contain the
    supplement (typically an amino acid;

   These reverse mutations are typically point mutations or
    frame shifts.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Fundamental principles:
   Many test strains have features that make them more
    sensitive for the detection of mutations:
     Specific responsive DNA sequences at the reversion sites;
     Increased cell permeability to large molecules
     Lack of DNA repair or enhancement of error-prone DNA
       repair;
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Advantages:
   Fast;
   Cheap;
   Quick screening for micro-DNA damage;
   Extensive database/library of the effects of a very diverse
    array of chemicals;
   Generally has acceptable false positive/false negative levels
    (i.e. good sensitivity, specificity, precision and predictive
    value)
   Although many compounds that are positive in this test are
    mammalian carcinogens, the correlation is not absolute. It is
    dependent on chemical class and there are carcinogens
    that are notdetected by this test because they act through
    other, non-genotoxic mechanisms or mechanismsabsent in
    bacterial cells.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays


 Although many compounds that are positive in this test are
  mammalian carcinogens, the correlation is not absolute. It is
  dependent on chemical class and there are carcinogens that
  are not detected by this test because they act through other,
  non-genotoxic mechanisms or mechanismsabsent in bacterial
  cells.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Disadvantages:
   Utilizes prokaryotic cells, which differ from mammalian cells in
     such factors as uptake, metabolism, chromosome structure
     and DNA repair processes;
   Tests conducted in vitro generally require the use of an
     exogenous source of metabolic activation. In vitro metabolic
     activation systems cannot mimic entirely the mammalian in
     vivo conditions;
   The test does not provide direct information on the
    mutagenic and carcinogenic potency of a substance in
    mammals.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Limitations:


   Difficult to use with substances that are potent bacteriocides
    or bacteriostats (i.e. substances that block mitosis);

   Culture media are hydrophilic i.e. requires specialized
    techniques for highly lipophilic substances (e.g. petroleum
    distillates);

   Special techniques are required for gases, vapors or
    substances that evaporate at 37OC;
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Metabolic activation systems:
   The objective is to replicate at least some of the major
    biotransformation pathways in vitro;
   Classical system is liver S9 fraction:
      Rats are dosed with Arochlor 1254 (a PCB mixture that acts as a
       potent inducer of liver CYP and UGT enzymes via the AhR
       pathway)  livers are homogenized  centrifuged at 9000 g for
       20 minutes  supernatant is collected;
      S9 contains cytosol and microsomes (= smooth endoplasmic
       reticulum): microsomes component contains cytochrome P450
       isoforms (phase I metabolism);cytosolic portion contains the
       major part of the activities of transferases (phase II metabolism)
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Metabolic activation systems:
   Classical system is liver S9 fraction:


      A NADPH-regenerating system or NADPH solution is required to
       supply the energy demand of the CYP enzymes (powers the CYP
       cycle);


      For the catalytic activity of phase II enzymes, addition of
       exogenous cofactors is necessary: UDPGA and alamethicin for
       UGT; acetyl CoA, DTT, and acetyl CoA regenerating g system for
       NAT; PAPS for ST; and GT for GST;
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Bacteria and strains required for OECD 471:
   Salmonella typhimurium TA1535 rfa+ uvrB+ hisG46:
   S. typhimurium TA1537 rfa+ uvrB+ hisC3076;
   S. typhimurium TA98 rfa+ uvrB+ hisD3052 pKM101;
   S. typhimurium TA100 rfa+ uvrB+ hisG46 pKM101;
   Escherichia coli WP2 trp+ uvrA;
   In order to detect cross-linking mutagens it may be preferable to
    include TA102 or to add a DNA repair-proficient strain of E.coli [e.g.
    E.coli WP2 or E.coli WP2 (pKM101)]
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Bacteria and strains required for OECD 471:So what does this all
  mean?
   His genotype: location of the deletion mutation in the histidine
    operon (operon is a functioning unit of genomic DNA containing a
    cluster of genes under the control of a single regulatory signal or
    promoter. Net result is that the bacterial carrying this mutation
    cannot synthesize histidine and are auxotrophs);


   rfa mutation: A mutation (rfa) in all strains that leads to a defective
    lipopolysaccharide (LPS) layer that coats the bacterial surface,
    making the bacteria more permeable to bulky chemicals;


   uvrB and uvrA mutations: The uvrB deletion mutation eliminates the
    accurate excision repair mechanism, thereby allowing more DNA
    lesions to be repaired by the error-prone DNA repairmechanism.
    The deletion through the biotin gene makes the bacteria biotin
    dependent.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Bacteria and strains required for OECD 471:So what does this all
  mean?
   Plasmid pKM101: present in strains TA1535 and TA1538 resulting in
    the corresponding isogenic strains TA100, TA98, TA97, TA102 and
    TA104. Plasmid pKM101 enhances chemical and UV-induced
    mutagenesis via an increase in the recombination DNA repair
    pathway. The plasmid confers ampicillin resistance, which is a
    convenient marker to detect the presence of the plasmid;


   Insertion of the mutation hisG428 on the multi-copy plasmid pAQl
    which was introduced in strain TA102 with the aim of amplifying the
    number of target sites. To enhance the ability of this strain to detect
    DNA crosslinking agents, the uvrB gene was retained making the
    bacterium DNA repair proficient
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Bacteria and strains required for OECD 471: So what does this all
  mean?
   trp+: Trp operon is that codes for the production of tryptophan. trp+
    E. coli are auxotrophs for tryptophan;


   Note: all strains except S. typhimurium TA102 are also biotin
    dependent (i.e. are both histidine and biotin auxotrophs).
E. Coli Wp2 uvrA:
Notes:
• A trace of histidine (or
  tryptophan
  depending on the
  auxotroph) + biotin
  are incorporated to
  allow for 1 or 2 rounds
  of cell replication in
  order to fix any
  mutations present;

• The plate
  incorporation method
  is reputed to increase
  the assay sensitivity
  and to allow the
  testing of suspensions
  as well as solutions of
  test article
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Modifications to the standard plate incorporation assay:

   The preincubation assay: the tester strains are exposed to the
    chemical for a short time (20 to 30min) in a small volume (0.5ml) of
    either buffer or S-9 mix, prior to plating on glucose agar minimal
    medium (GM agar) supplemented with a trace amount of histidine.
    With few exceptions it is believed that this assay is more sensitive
    than the plate incorporation assay, because short-lived mutagenic
    metabolites may have a better chance reacting with the tester
    strains in the small volume of preincubation mixture, and the
    effective concentration of S-9 mix in the preincubation volume is
    higher than that on the plate.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Modifications to the standard plate incorporation assay:

   The desiccator assay for liquids and gases: the use of a closed
    chamber is recommended for testing highly volatile chemicals and
    gases;
   The Kado Salmonella microsuspension assay for testing samples of
    small volumes;


   Testing chemicals in a reduced oxygen atmosphere: anaerobic
    environments, such as anaerobic chambers, have been used to
    study mutagenicity of chemicals and fecal samples under reduced
    oxygen levels.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Modifications to the standard plate incorporation assay:

   Fluctuation method: The fluctuation method is performed entirely in liquid
    culture and is scored by counting the number of wells that turn yellow from
    purple in a 96-well microplate. If bacteria are able to revert back to
    metabolic competence they will continue to replicate and turn the liquid
    media acid. By including a pH indicator in the media, the frequency of
    mutation is counted as the number of wells out of 96 which have changed
    color.


   The fluctuation method is comparable to the traditional pour plate method
    in terms of sensitivity and accuracy, however, it does have a number of
    advantages, namely, allowing for the testing of higher concentrations of
    sample (up to 75% v/v), increasing the sensitivity and extending its
    application to low-level environmental mutagens.[20]


   The fluctuation method also has a simple colorimetric endpoint; counting
    the number of positive wells out of a possible 96 wells is much less time
    consuming than counting individual colonies on an agar plate.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Strain checks:
   Histidine dependence (his): streak a loopful of the culture across a
    GM agar plate supplemented with an excess of biotin. Because all
    the Salmonella strains are histidine dependent, there should be no
    growth on the plates.


   Biotin dependence (bio): streak a loopful of the culture across a
    GM agar plate supplemented with an excess of histidine. There
    should be no growth on the plate except for strain TA102 which is
    biotin independent.
   Biotin and histidine dependence (bio, his): streak a loopful of the
    culture across a GM agar plate supplemented with an excess of
    biotin and histidine. Growth should be observed with all strains.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Strain checks:
   rfa marker: streak a loopful of the culture across a GM agar plate
    supplemented with an excess of biotin and histidine. Apply 10 µl of
    a sterile 0.1% crystal violet solution. All Salmonella strains should
    show a zone of growth inhibition (crystal violet is a relatively large
    bacteriocidal molecule [mw = 407.979] which cannot penetrate
    the bacteria if a normal cell wall is present);


   Presence of plasmid pKM101 (ampicilline resistance): apply in the
    center of a plate 10 µl of ampicilline solution. Streak a loopful of the
    pKM101-carrying Salmonella culture across an agar plate
    supplemented with an excess of histidine and biotin. Growth should
    be observed.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Strain checks:
   Spontaneous mutant frequency: use the standard plate
    incorporation assay procedure without the inclusion of a solvent for
    determining the spontaneous mutant frequency (negative control)
    of each of the tester strains. When the spontaneous control values
    fall outside an acceptable range the genetic integrity of the strain is
    considered compromised, and a new culture should be isolated.
Classical Assays for Genotoxicity:
        Bacterial reverse mutation assays
 Controls:
   Must have: suitable positive control for non-metabolic activation
    and metabolic activation + solvent/vehicle negative control;
For tests with metabolic activation:
Classical Assays for Genotoxicity:
         Bacterial reverse mutation assays
 Evaluation of results:
   Surviving populations: usually a 2 – 3-log range of doses are used
    and the highest of these doses is selected to show some degree of
    bacterial toxicity (background clearing or reduction in the number
    of spontaneous mutants);
   Dose-response phenomena: a mutagen should display a clear
    dose-related increase in revertant colonies (can be influenced by
    poor dose range selection);
Classical Assays for Genotoxicity:
         Bacterial reverse mutation assays
 Evaluation of results:


   Generally speaking a mutagen will produce a positive dose
    response over at least 3 different concentrations with the hiighest
    increase in revertants being 2 – 3 times that of the level of
    spontaneous revertants in the negative control plates;


   Pattern: TA-1535 and TA-100 are derived from the same parental
    strain, thus commonly the responses of these two strains should be
    nearly identical;


   The results (particularly the pattern of reversion) should be
    repeatable and consistent;


   Specific gene sequencing may be of use in some cases.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Basic principle of the assay:


   Cells deficient in thymidine kinase (TK) due to the mutation
    TK+/- → TK-/- are resistant to the cytotoxic effects of the
    pyrimidine analogue trifluorothymidine (TFT);

   TFT is converted to TFT-monophosphate by thymidine kinase
     TFT-triphosphate (TFT-TP) is incorporated into DNA, resulting
    in cytocidal effects.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Basic principle of the assay:

   Thymidine kinase proficient cells are sensitive to TFT, which
    causes the inhibition of cellular metabolism and halts further
    cell division;

   Thus mutant cells are able to proliferate in the presence of
    TFT, whereas normal cells, which contain thymidine kinase,
    are not.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Basic principle of the assay:

   The TK mutations have no effect on the growth of cells in
    normal media because normal DNA synthesis does not
    involve the TK pathway;

   The L5178Y cells also have point mutations in both p53
    alleles,. Because p53 is an important protein in the DNA
    damage response in the cell, the L5178Y cell line is
    inadequate in its response to DNA damage, which is
    arguably important for enhanced assay sensitivity for the
    detection of genotoxic compounds.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Basic principle of the assay:
   Assay is usually conducted with and without S9 metabolic
    activation;

 Controls:
   Vehicle/solvent negative control;
   Positive controls: methylmethanesulfonate for studies without
    metabolic activation; cyclophosphamide, benzo(a)pyrene
    and 3-methylcholanthrene for tests with metabolic
    activation;
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Dose range:
   Selected so that the test doses span the range from 0% to
    80% reduction in cell growth with or without S9 activation.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Assay acceptance criteria:
   The average absolute cloning efficiency must be in the
     range of 65 – 120% (ability of single cells to form a new
     colony);

   The average increase in the vehicle control cell population
    should be 8 -32 fold over 2 days;

   Background forward mutation frequency should be
    acceptable;
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Assay acceptance criteria:
   The frequency of forward mutation in the positive controls
     must be consistent with historical data for the lab;

   The assay must include applied concentrations that reach 5
    mg/mL or 10 mM (which ever is the lower) for test articles that
    cause little or no cytotoxicity;

   The assay must include applied concentrations that reduce
    the relative cell growth by approximately 20%; OR

   Reach a concentration that exceeds the solubility limit.
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Interpretation:
   Cell colony size on agar: large mutant colonies have few
     genetic changes other than the tk+/-  tk-/- forward mutation;
     small mutant colonies generally represent more extensive
     genetic modification of the cells;

   A mutant frequency of at least 2 times that in the negative
    control is suggestive of a positive control (induction of
    mutation is an additive process and not a multiple over
    background – the global evaluation factor should be used);
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Interpretation:
   Global evaluation factor = the mean of each vehicle control
     mutant frequency distribution + 1 SD across multiple test labs;
   The GEF for the agar assay method = 90;
   The GEF for 96 well plate assays = 126;

   A positive assay is one where the mutant frequency for the
    test article equals or exceeds the GEF PLUS there is a
    statistically demonstrable dose trend present;

   A negative assay is one where the mutant frequency for the
    test article is below the GEF PLUS there is no statistically
    demonstrable dose trend present
Classical Assays for Genotoxicity:
           Mouse lymphoma L5178Y tk+/- forward
           mutation assay (OECD 476)
 Interpretation:
   If only one criterion is met, additional studies are needed to
     clarify the test outcome
Classical Assays for Genotoxicity:
           Other mammalian cell forward
           mutation tests(OECD 476)

 The HRPT assay:
   Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) is
    a transferase that catalyzes conversion of hypoxanthine to
    inosine monophosphate and guanine to guanosine
    monophosphate;

   Test cell lines are HRPT+/- i.e. have a single functional HRPT
    allele that is lost with mutation;

   Multiple different mammalian cell lines with this genotype
    are available: V79 Chinese hamster cells, AS52 Chinese
    hamster cells, Chinese hamster ovary cells (CHO) and human
    TK6 human lymphoblastoid cells.
Classical Assays for Genotoxicity:
          Other mammalian cell forward
          mutation tests(OECD 476)

 The HRPT assay:
   Basic principles: HRPT converts 6-thioguanine (TG) or 8-
    azaguanine (AG) to non-toxic metabolites;

   Cells that have HRPT function are able to survive and
    replicate in the presence of TG or AG;

   Those cells that lack HRPT function due to a forward mutation
    die in the presence of TG or AG;

   The assay is conducted and assessed in a manner that is
    similar to the TK assay.
Classical Assays for Genotoxicity:
          Other mammalian cell forward
          mutation tests(OECD 476)

 The XHRPT assay:


   Transgene of xanthineguanine phosphoribosyl transferase
    (XHRPT);
   Works on the same principle as the HRPT assay;
   XHRPT is located on autosomal chromosomes where as HRPT
    is located on the X chromosome.
Classical Assays for Genotoxicity:
           Other mammalian cell forward
           mutation tests(OECD 476)



 The TK, HPRT and XPRT mutation tests detect different spectra of
  genetic events. The autosomal location of TK and XPRT may
  allow the detection of genetic events (e.g. large deletions) not
  detected at the HPRT locus on X-chromosomes.
Classical Assays for Genotoxicity:
          In vivo mammalian forward mutation
          assays in transgenic animals.

 The Big Blue mouse assay:
   The genome of these mice has been manipulated such
    that every cell contains, stably integrated into the
    DNA, multiple tandem copies of a bacterial lac I repressor
    gene;

   lf the mice are exposed to mutagens, there is a small
    probability that a mutation will occur somewhere along
    the inserted sequence;

   Any mutation will lead to an inactive lac I gene and lac
    repressor protein, meaning the gene (lacZ) for beta-
    galactosidase will no longer be repressed;
Classical Assays for Genotoxicity:
          In vivo mammalian forward mutation
          assays in transgenic animals.

 The Big Blue mouse assay:
   The DNA is extracted from the tissues of the treated mouse 
    the vector is isolated and used to make functional
    bacteriophages  E. coli cells are mixed with the
    bacteriophage and spread on a solid culture medium  the
    bacteriophages infect and destroy ("lyze") the E. coli cells --?
    this causes clear circular zones, called plaques, to appear in
    a "lawn" of bacteria  Before they die, cells that have been
    infected by bacteriophages carrying a mutated lac I will
    produce beta-galactosidase  This reacts with a substrate in
    the culture medium turning it blue Count both colorless
    and blue plaques  The number of blue plaques divided by
    the total number of plaques gives the mutation frequency.
Classical Assays for Genotoxicity:
          In vivo mammalian forward mutation
          assays in transgenic animals.

 The Big Blue mouse assay:
   Bacteriophages with non-mutated genes produce colorless
    plaques because no beta-galactosidase is synthesized;
Classical Assays for Genotoxicity:
          In vivo mammalian forward mutation
          assays in transgenic animals.



 The muta-mouse assay operates in a manner similar to the
  Big Blue except it uses a LacZ gene
Classical Assays for Genotoxicity:
           In vivo mammalian forward mutation
           assays in transgenic animals.

 In vivo forward mutation assays offer very substantial
  advantages:
   The complete array of metabolic processes are present;
   Tissue specific metabolism is taken into account rather than
     just the over simplified metabolism that occurs with S9;
   Tissue and even cell-type specific mutation rates can be
     measured. This includes germ cell (spermatogonial)
     mutations in males!;
   Takes into account toxicokinetic and toxicodynamic
     differences between different organs, tissues and cell types;
   In general, tissue specific mutation frequencies generally
    match the distribution of mutation in live animals!
Classical Assays for Genotoxicity:
           In vitro chromosomal aberration
     assay
           (OECD 473)
 Basic principle of the assay:
   Typically Chinese hamster ovary cells strain CHO-WBL ATCC
    CCL61 are used (fibroblastic cell line);

      Chromosomal number of 21 with a low frequency of
       spontaneous mutations;

   Alternatively human peripheral blood lymphocytes that have
    been stimulated to divide using a mitogen (PHA) are used;
Classical Assays for Genotoxicity:
           In vitro chromosomal aberration
     assay
           (OECD 473)
 Basic principle of the assay:
   Cell cultures are exposed to the test substance both with
    and without metabolic activation

   At predetermined intervals after exposure of cell cultures to
    the test substance, they are treated with a metaphase-
    arresting substance (e.g. Colcemid® or colchicine),
    harvested, stained and metaphase

   Cells are analysed microscopically for the presence of
    chromosome aberrations.
Classical Assays for Genotoxicity:
           In vitro chromosomal aberration
     assay
           (OECD 473)
 Basic principle of the assay:
   Cell cultures are exposed to the test substance both with
    and without metabolic activation

   At predetermined intervals after exposure of cell cultures to
    the test substance, they are treated with a metaphase-
    arresting substance (e.g. Colcemid® or
    colchicine), harvested, stained and metaphase

   Cells are analysed microscopically for the presence of
    chromosome aberrations.
Classical Assays for Genotoxicity:
           In vitro chromosomal aberration
     assay
           (OECD 473)
 Measurement of results:
   This assay does NOT determine aneuploidy (i.e. increased or
    decreased number of chromosomes; this is because of
    artifacts associated with the cell analysis preparation): only
    cells with a normal chromosome number are analysed;
   The types of chromosomal aberrations that are detected
    are:
     Simple breaks;
     Complex exchanges;
     Gaps;
     Dicentric chromosomes;
     Ring chromosomes.
Classical Assays for Genotoxicity:
           In vitro chromosomal aberration
     assay
           (OECD 473)
 Advantages:
   Accurate identification of all the different chromosome
    mutation types;
   Possible co-detection of mitotic indices;
   No full automatic but interactive scoring possible;

 Disadvantages:
   High false positive rate;
   Labor intensive and time consuming;
   Heavily dependent on operator/reader skill.
Classical Assays for Genotoxicity:
           In vivo bone marrow chromosomal
           aberration assay (OECD 475)

 Advantages over the in vitro method:
   Takes into account in vivo TK and TD to a certain extent;

   Can test different routes of exposure;

   Full in vivo metabolism (but with some limitations, particularly
    given that the tissue analyzed is bone marrow);
Classical Assays for Genotoxicity:
          In vivo bone marrow chromosomal
          aberration assay (OECD 475)

 Disadvantages over the in vitro method:
   Exposure of the bone marrow must occur in order for the test
    to be valid – this may need to be demonstrated by a TK
    study;


   If there is evidence that the test substance, or a reactive
    metabolite, will not reach the targettissue, it is not
    appropriate to use this test;
Classical Assays for Genotoxicity:
          In vivo bone marrow chromosomal
          aberration assay (OECD 475)

 Disadvantages over the in vitro method:

   If metabolism to an ultimate mutagen is required, then the
    blood and tissue T½ must be long enough for bone marrow
    exposure to occur unless local metabolism in the bone
    marrow occurs;

   Risk of excessive toxicity producing distorted results: dose
    ranging studies are often needed.
Classical Assays for Genotoxicity:
           In vivo bone marrow chromosomal
           aberration assay (OECD 475)

 Principle of the assay:
   Animals (typically rats or Chinese hamsters) are exposed to
    the test substance by an appropriate route of exposure and
    are sacrificed at appropriate times after treatment;

   Prior to sacrifice, animals are treated with a metaphase-
    arresting agent (e.g., colchicine or Colcemid®);

   Chromosome preparations are then made from the bone
    marrow cells and stained, and metaphase cells are analysed
    for chromosome aberrations.
Classical Assays for Genotoxicity:
           In vivo spermatogonial chromosomal
           aberration assay (OECD 483)

 Assay is similar in principle to OECD 475;

 Important difference is that it tests for GERM CELL
  chromosomal damage;

 If there is evidence that the test substance, or a reactive
  metabolite, will not reach the target tissue, it is not
  appropriate to use this assay.
Classical Assays for Genotoxicity:
          Unscheduled DNA Synthesis OECD428

 This is essentially a test of DNA repair that occurs
  outside of the normal period of DNA synthesis in cells;

 Measures DNA synthesis in cells which are not in the S
  phase of the cell cycle;

 The assay measures global genomic nucleotide
  excision repair (NER);
Classical Assays for Genotoxicity:
          Unscheduled DNA Synthesis OECD428

 The classical OECD assay measures DNA synthesis by
  measuring the incorporation of 3H-thymidine or BrDU
  into DNA;

 More modern techniques use specific DNA dyes and
  flow cytometry: faster, more accurate, provides
  more information;
Classical Assays for Genotoxicity:
           Unscheduled DNA Synthesis OECD428

 The test requirements are very broad:
   Can be done in vitro or in vivo;
   Primary cell cultures or established cell lines can be used;
   Test is performed with or without metabolic activation;
   In order to discriminate between UDS and normal semi-
    conservative DNA replication, cell replication is inhibited or
    minimized using an arginine-deficient medium, low serum
    content, or by hydroxyurea in the culture medium;
   For flow cytometric methods, inhibition of the cell cycle is not
    required.
Classical Assays for Genotoxicity:
           Unscheduled DNA Synthesis OECD428

 Given that the assay measures nucleotide excision repair, it is
  critical that:
   At least some of the DNA lesions produced by the test article
      are repairable by nucleotide excision repair;
   The test cell line/animal line is capable of relatively normal
      nucleotide excision repair
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Basic principle of the assay:
   It is an assay of macro-chromosomal damage (chromosome
    fragments lacking a kinetocore/centromere) OR damage to
    the mitotic spindle (whole chromosomes chromosome
    fragments containing a kinetocore);
   The assay detects lagging chromosome fragments or
    lagging chromosomes;
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Basic principle of the assay:
   Takes advantage of the fact that when bone marrow
    erythroblast develops into a polychromatic erythrocyte the
    cell nucleus is extruded from the cell however DNA
    containing micronuclei are not extruded from the cell;

   Micronuclei can be detected by cell staining and flow
    cytometry. The lack of a normal cell nucleus makes the
    detection of micronuclei much easier.
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474




 This assay is regarded as the highest tier assay of the commonly
  conducted genetic toxicology assays for chemicals and other
  agents;
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Important features:
   Classically outbred rats and mice are used in preference to
    inbred strains in order to reduce the likelihood of strain
    specific responses (classically CD-1 [ICR] BR mice and/or
    Sprague-Dawley CD [SD] IGS BR rats);
   Animals are usually 8 -10 weeks old at the start of the study;
   Various routes of administration, including parenteral routes,
    are available. Unless there are specific reasons not to, the
    study route of exposure should match those of the likely
    human routes of exposure;
   Parenteral routes (IV, IP) are used if absorption is poor or if
    there are other technical constraints regarding dosing.
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Important features:
   The IV route of exposure has a particular advantage in that it
    guarantees exposure of the bone marrow;

   If non-IV routes of exposure are used, there must be sufficient
    TK data that demonstrates that bone marrow exposure
    occurs or is likely to occur;

   If the test article is a pro-mutagen (i.e. requires metabolic
    activation), then there must be reasonable certainty that
    appropriate metabolic activation can occur in the bone
    marrow OR the active metabolites have a chemical half-life
    that is long enough for them to reach the bone marrow!
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Important features:
   Ideally, 2 positive controls should be used:
     Cyclophosphamide: requires metabolic activation;
     A substance that does not require metabolic activation
       (e.g. ethyl methanesulfonate)
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Positive controls:
   Can be administered by a different route than the test
    article;
   Can be administered as a single dose;

 Negative controls: typically a solvent or vehicle control
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 “No standard treatment schedule (i.e. 1, 2, or more treatments
  at 24 h intervals) can be recommended. The samples from
  extended dose regimens are acceptable as long as a positive
  effect has been demonstrated for this study or, for a negative
  study, as long as toxicity has been demonstrated or the limit
  dose has been used, and dosing continued until the time of
  sampling. Test substances may also be administered as a split
  dose, i.e., two treatments on the same day separated by no
  more than a few hours, to facilitate administering a large
  volume of material.”
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 The test can be performed in 2 ways:

 “Animals are treated with the test substance once. Samples of
  bone marrow are taken at least twice, starting not earlier than
  24 hours after treatment, but not extending beyond 48 hours
  after treatment with appropriate interval(s) between samples.
  The use of sampling times earlier than 24 hours after treatment
  should be justified. Samples of peripheral blood are taken at
  least twice, starting not earlier than 36 hours after treatment,
  with appropriate intervals following the first sample, but not
  extending beyond 72 hours. When a positive response is
  recognized at one sampling time, additional sampling is not
  required.”
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 The test can be performed in 2 ways:

 “If 2 or more daily treatments are used (e.g. two or more
  treatments at 24 hour intervals), samples should be collected
  once between 18 and 24 hours following the final treatment for
  the bone marrow and once between 36 and 48 hours following
  the final treatment for the peripheral blood (12).”
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Dose rates:
   “If a range finding study is performed because there are no suitable
    data available, it should be performed in the same laboratory,
    using the same species, strain, sex, and treatment regimen to be
    used in the main study (13). If there is toxicity, three dose levels are
    used for the first sampling time. These dose levels should cover a
    range from the maximum to little or no toxicity. At the later
    sampling time only the highest dose needs to be used. The highest
    dose is defined as the dose producing signs of toxicity such that
    higher dose levels, based on the same dosing regimen, would be
    expected to produce lethality. “


   Toxicity can take any form. A commonly used measure is a
    reduction in the ratio of polychromatic to normochromatic
    erythrocytes (what does this say about the bone marrow?);
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Dose rates:
   “Substances with specific biological activities at low non-toxic doses
    (such as hormones and mitogens) may be exceptions to the dose-
    setting criteria and should be evaluated on a case-by-case basis.
    The highest dose may also be defined as a dose that produces
    some indication of toxicity of the bone marrow (e.g. a reduction in
    the proportion of immature erythrocytes among total erythrocytes
    in the bone marrow or peripheral blood).”


   In general, a maximum limit dose of 2 g/kg is used for substances
    with low toxicity
Classical Assays for Genotoxicity:
           Mammalian Erythrocyte Micronucleus
           Test OECD 474

 Sexes:
   In general, if there is data that indicates that there are no sex
    differences in toxicity, only males are used;

   If there is data indicating sex differences, BOTH sexes MUST
    be used.
Classical Assays for Genotoxicity:
          Mammalian Erythrocyte Micronucleus
          Test OECD 474

 Assay acceptance criteria:
   % micronucleated PCE in the negative control group must lie
    within the historical normal range for the lab and must be
    below 0.4%;

   Must be a statistically significant increase in the %
    micronucleated PCE relative to the negative controls in the
    positive control group and this increase should be consisted
    with historical data for the lab;

   Either the limit dose is reached or at least some non-leathal
    toxicity must be present in the highest test article dose
Classical Assays for Genotoxicity:
          Mammalian Erythrocyte Micronucleus
          Test OECD 474

 What constitutes a positive response?
   A statistically significant increase in micronucleated
    PCE in at least one of the test article doses relative to
    the negative controls; OR

   A statistically significant dose response relationship is
    present;

   The results make biological sense.
Classical Assays for Genotoxicity:
          Rare Assays

 Mouse Spot Test (OECD 484)
   Now extremely difficult to perform because the appropriate
    mouse strains are no longer available (lost);
   It is a high tier assay in terms of risk assessment/hazard
    classification;
   Often now replaced by Big Blue or Mutamouse
   An in vivo test in mice in which developing embryos are
    exposed to a test agent.
   The target cells are melanoblasts;
   The target genes are those which control the pigmentation
    of the coat hairs;
Classical Assays for Genotoxicity:
           Rare Assays

 Mouse Spot Test (OECD 484)
   The developing embryos are heterozygous for a number of these
    coat colour genes;


   A mutation in, or loss of (by a variety of genetic events), the
    dominant allele of such a gene in a melanoblast results in the
    expression of the recessive phenotype in its descendant
    cells, constituting a spot of changed colour in the coat of the
    resulting mouse.


   The number of offspring with these spots, mutations, are scored and
    their frequency is compared with that among offspring resulting
    from embryos treated with the solvent only. The mouse spot test
    detects presumed somatic mutations in foetal cells.
Figure 2Defects in melanocyte development cause white spotting, while the
stem cell defect results hair graying.

(A) A Ednrbs-l/Ednrbs-l mouse demonstrating extensive piebald spotting. (B) A
KitW-2J/+ mouse demonstrating a white head blaze, and small dorsal spot on the
back. (C) A Sox10Lacz/+ mouse exhibiting the characteristic white belly spot. (D)
A Mitfvit/vit mouse (upper) exhibits gradual hair graying. A lower mouse is age-
matched control. Adapted from the WEB site of the European Society for
Pigment Cell Research (ESPCR), “Color genes”: http://www.espcr.org/micemut/.
Classical Assays for Genotoxicity:
          Rare Assays

 Mouse Heritable Translocation Assay (OECD 485)
   This is a particularly useful assay because it is an in vivo
    measure of germ cell macro-chromosomal damage;
   Currently there is no commonly used in vivo equivalent to this
    assay;
   There are 2 forms of the assay:
     Detection of changes in fertility of the F1 progeny of
       exposed animals;
     Cytogenetic examination of the F1 progeny of exposed
       animals;
Classical Assays for Genotoxicity:
           Rare Assays

 Mouse Heritable Translocation Assay (OECD 485)
   The assay primarily measures the frequency of structural
    chromosome assays that do not result in a loss of
    genetic material i.e. reciprocal translocations;

   Reciprocal translocations = fetal survival + no
    malformations;

   Non-reciprocal translocations usually mean fetal death;
Classical Assays for Genotoxicity:
           Rare Assays

 Mouse Heritable Translocation Assay (OECD 485)
   Reciprocal translocation, however, result in a loss of fertility in
    the F1 generation due to the production of unbalanced
    chromosomes in the gametes (i.e. unbalanced
    translocations);
Classical Assays for Genotoxicity:
          Rare Assays

 Insect and yeast OECD assays: rarely if ever used. You will
  occasionally come across them in historical data sets.

                  Treat them with caution!
Other Common Genotoxicity Assays:
         The comet assay

 = Single Cell Electrophoresis Assay;

 Simple, reliable and particularly useful!;

 Well accepted assay;

 Can also be used as a very simple, but effective and
  accurate assay of DNA repair;
Other Common Genotoxicity Assays:
         The comet assay

 Principle:
   Measures DNA strand breaks (i.e. clastenogenesis);
   Can be done in vitro or in vivo;
   Can be used for specific tissues or even specific cells;
   Cells embedded in agarose on a microscope slide are lysed with
    detergent and high salt to form nucleoids containing supercoiled
    loops of DNA linked to the nuclear matrix;
   Electrophoresis at high pH results in structures resembling comets,
    observed by fluorescence microscopy;
   The intensity of the comet tail relative to the head reflects the
    number of DNA breaks
A Few Slides of Gratuitous Silyness


After 228 power point slides on genetic toxicology, I figured you
were entitled…..
Before Genetic Tox   After Genetic Tox
?

More Related Content

What's hot

Dh 111 radiation biology (ch.4) power point
Dh 111 radiation biology (ch.4) power pointDh 111 radiation biology (ch.4) power point
Dh 111 radiation biology (ch.4) power pointdinardh
 
Excision repair in dna
Excision repair in dnaExcision repair in dna
Excision repair in dnaIrene Daniel
 
Arsenic toxicity
Arsenic toxicityArsenic toxicity
Arsenic toxicityabha60
 
A Chiroptical Photoswitchable DNA Complex
A Chiroptical Photoswitchable DNA ComplexA Chiroptical Photoswitchable DNA Complex
A Chiroptical Photoswitchable DNA ComplexAngela Mammana
 
Synthesis and characterization of water-soluble free-base, zinc and copper
Synthesis and characterization of water-soluble free-base, zinc and copperSynthesis and characterization of water-soluble free-base, zinc and copper
Synthesis and characterization of water-soluble free-base, zinc and copperAngela Mammana
 
Sds presentation rockefeller[1][1]
Sds presentation rockefeller[1][1]Sds presentation rockefeller[1][1]
Sds presentation rockefeller[1][1]Dr Robert Craig PhD
 
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...RachaelWong11
 
Science Fair Presentation
Science  Fair  PresentationScience  Fair  Presentation
Science Fair Presentationguestb484e1
 
2008 OU Seminar
2008 OU Seminar2008 OU Seminar
2008 OU Seminarpipessvc
 
Fate of chlorophyll degradation
Fate of chlorophyll degradationFate of chlorophyll degradation
Fate of chlorophyll degradationAmalJutt1
 
Biologic effects Daniel jayaprakash
Biologic effects Daniel jayaprakashBiologic effects Daniel jayaprakash
Biologic effects Daniel jayaprakashalmasmkm
 

What's hot (20)

Dh 111 radiation biology (ch.4) power point
Dh 111 radiation biology (ch.4) power pointDh 111 radiation biology (ch.4) power point
Dh 111 radiation biology (ch.4) power point
 
Excision repair in dna
Excision repair in dnaExcision repair in dna
Excision repair in dna
 
Arsenic toxicity
Arsenic toxicityArsenic toxicity
Arsenic toxicity
 
Sos repair mechanism
Sos repair mechanismSos repair mechanism
Sos repair mechanism
 
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
 
DNA repair mechanism
DNA repair mechanism DNA repair mechanism
DNA repair mechanism
 
Daniel Young-Farhat 2016
Daniel Young-Farhat 2016Daniel Young-Farhat 2016
Daniel Young-Farhat 2016
 
Dna
DnaDna
Dna
 
A Chiroptical Photoswitchable DNA Complex
A Chiroptical Photoswitchable DNA ComplexA Chiroptical Photoswitchable DNA Complex
A Chiroptical Photoswitchable DNA Complex
 
Dna replication MBBS
Dna replication MBBSDna replication MBBS
Dna replication MBBS
 
opiod probe poster
opiod probe posteropiod probe poster
opiod probe poster
 
Synthesis and characterization of water-soluble free-base, zinc and copper
Synthesis and characterization of water-soluble free-base, zinc and copperSynthesis and characterization of water-soluble free-base, zinc and copper
Synthesis and characterization of water-soluble free-base, zinc and copper
 
Structure of DNA
Structure of DNA Structure of DNA
Structure of DNA
 
Sds presentation rockefeller[1][1]
Sds presentation rockefeller[1][1]Sds presentation rockefeller[1][1]
Sds presentation rockefeller[1][1]
 
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
 
Science Fair Presentation
Science  Fair  PresentationScience  Fair  Presentation
Science Fair Presentation
 
2008 OU Seminar
2008 OU Seminar2008 OU Seminar
2008 OU Seminar
 
Fate of chlorophyll degradation
Fate of chlorophyll degradationFate of chlorophyll degradation
Fate of chlorophyll degradation
 
Biologic effects Daniel jayaprakash
Biologic effects Daniel jayaprakashBiologic effects Daniel jayaprakash
Biologic effects Daniel jayaprakash
 
Aadils ppt
Aadils pptAadils ppt
Aadils ppt
 

Viewers also liked

Proximal renal tubule physiology
Proximal renal tubule physiology Proximal renal tubule physiology
Proximal renal tubule physiology Ahad Lodhi
 
Micro rna diagnostics and therapeutics in acute kidney injury
Micro rna diagnostics and therapeutics in acute kidney injuryMicro rna diagnostics and therapeutics in acute kidney injury
Micro rna diagnostics and therapeutics in acute kidney injuryChristos Argyropoulos
 

Viewers also liked (20)

Stomach liver tumors-journal_club
Stomach liver tumors-journal_clubStomach liver tumors-journal_club
Stomach liver tumors-journal_club
 
Uof q2011final
Uof q2011finalUof q2011final
Uof q2011final
 
Thyroid
ThyroidThyroid
Thyroid
 
Nicnas carcinogenesis9
Nicnas carcinogenesis9Nicnas carcinogenesis9
Nicnas carcinogenesis9
 
Growth body weight-feed
Growth body weight-feedGrowth body weight-feed
Growth body weight-feed
 
Ttc cope 2
Ttc cope 2Ttc cope 2
Ttc cope 2
 
Jc liver tumors
Jc liver tumorsJc liver tumors
Jc liver tumors
 
Role of toxicology in regulatory processes 1
Role of toxicology in regulatory processes 1Role of toxicology in regulatory processes 1
Role of toxicology in regulatory processes 1
 
Renal nicnas-2012
Renal nicnas-2012Renal nicnas-2012
Renal nicnas-2012
 
The erythron
The erythronThe erythron
The erythron
 
The leukon
The leukonThe leukon
The leukon
 
Fabry Disease Urinary Podocyte Loss - 14 February 2014
Fabry Disease Urinary Podocyte Loss - 14 February 2014Fabry Disease Urinary Podocyte Loss - 14 February 2014
Fabry Disease Urinary Podocyte Loss - 14 February 2014
 
Proximal renal tubule physiology
Proximal renal tubule physiology Proximal renal tubule physiology
Proximal renal tubule physiology
 
Fiber toxicology
Fiber toxicologyFiber toxicology
Fiber toxicology
 
Tribal directory 2014
Tribal directory 2014 Tribal directory 2014
Tribal directory 2014
 
Robert T. Dunn, II, Ph.D., DABT, SLAS ADMET Special Interest Group Meeting p...
 Robert T. Dunn, II, Ph.D., DABT, SLAS ADMET Special Interest Group Meeting p... Robert T. Dunn, II, Ph.D., DABT, SLAS ADMET Special Interest Group Meeting p...
Robert T. Dunn, II, Ph.D., DABT, SLAS ADMET Special Interest Group Meeting p...
 
Liver nicnas-nov-2012
Liver nicnas-nov-2012Liver nicnas-nov-2012
Liver nicnas-nov-2012
 
Renal nicnas-2012 copy
Renal nicnas-2012 copyRenal nicnas-2012 copy
Renal nicnas-2012 copy
 
Micro rna diagnostics and therapeutics in acute kidney injury
Micro rna diagnostics and therapeutics in acute kidney injuryMicro rna diagnostics and therapeutics in acute kidney injury
Micro rna diagnostics and therapeutics in acute kidney injury
 
Dermal toxicology
Dermal toxicologyDermal toxicology
Dermal toxicology
 

Similar to Nicnas gen tox3 (20)

Drug dna interaction
Drug dna interactionDrug dna interaction
Drug dna interaction
 
DNA reparing
DNA reparingDNA reparing
DNA reparing
 
Project Ppt
Project PptProject Ppt
Project Ppt
 
Medicinal chemistry of anticancer drugs ppt
Medicinal chemistry of anticancer drugs pptMedicinal chemistry of anticancer drugs ppt
Medicinal chemistry of anticancer drugs ppt
 
DNA CHEMISTRY
DNA CHEMISTRYDNA CHEMISTRY
DNA CHEMISTRY
 
DNA REPAIR.pptx
DNA REPAIR.pptxDNA REPAIR.pptx
DNA REPAIR.pptx
 
DNA Damage and DNA Repair- Dr. Sonia Angeline
DNA Damage and DNA Repair- Dr. Sonia AngelineDNA Damage and DNA Repair- Dr. Sonia Angeline
DNA Damage and DNA Repair- Dr. Sonia Angeline
 
Dna damage repair
Dna damage repairDna damage repair
Dna damage repair
 
MUTATION.pdf
MUTATION.pdfMUTATION.pdf
MUTATION.pdf
 
Dna damage
Dna damageDna damage
Dna damage
 
DNA damage, types by kk sahu
DNA damage, types by kk sahuDNA damage, types by kk sahu
DNA damage, types by kk sahu
 
DNA damage and DNA repair
DNA damage and DNA repairDNA damage and DNA repair
DNA damage and DNA repair
 
Nucleic acids
Nucleic acids   Nucleic acids
Nucleic acids
 
DNA Repair .pptx
DNA Repair .pptxDNA Repair .pptx
DNA Repair .pptx
 
Molecular basis of tumor
Molecular basis of tumorMolecular basis of tumor
Molecular basis of tumor
 
Microbial genetics microbiology ar
Microbial genetics microbiology arMicrobial genetics microbiology ar
Microbial genetics microbiology ar
 
Molecular basis of mutations
Molecular basis of mutationsMolecular basis of mutations
Molecular basis of mutations
 
DNA structure 2.pptx molecular biology.pptx
DNA structure 2.pptx molecular biology.pptxDNA structure 2.pptx molecular biology.pptx
DNA structure 2.pptx molecular biology.pptx
 
IDENTIFICATION OF PROTEIN BINDING SITE.docx
IDENTIFICATION OF PROTEIN BINDING SITE.docxIDENTIFICATION OF PROTEIN BINDING SITE.docx
IDENTIFICATION OF PROTEIN BINDING SITE.docx
 
Mutation
MutationMutation
Mutation
 

Nicnas gen tox3

  • 1. Genetic Toxicology Dr R B Cope BVSc BSc(Hon 1) PhD cGLPCP DABT ERT
  • 2. Quick Review of Relevant Basic Biology
  • 3.
  • 4.
  • 5.
  • 6.
  • 7.
  • 8.
  • 9.
  • 10.
  • 11.
  • 12.
  • 13.
  • 14.
  • 15.
  • 16. Definitions  Genotoxic:  Non-genotoxic:  Direct-acting mutagen:  Indirect-acting mutagen:  Pro-mutagen:  Proximate mutagen:
  • 17. Definitions  Ultimate mutagen:  Alkylating agent:  Radiomimetic:  Non-radiomimetic:  DNA macrolesion  DNA microlesion
  • 18. Direct-acting mutagens  By definition, these are highly DNA reactive agents that do not require prior metabolism for direct DNA interactions;  Generally very reactive chemicals, often with relatively short environmental half-lives;  Tend to be highly reactive electrophiles;  Typically act at the site of first contact;
  • 19. Direct-acting mutagens  Chemical stability, transport, membrane permeability generally determine the degree of mutagenicity associated with these agents;  Direct-acting mutagens are generally mutagenic at multiple tissue sites and in essentially all species;
  • 20. Direct-acting mutagens  Classical examples:  Nitrogen and sulfur mustards;  Methyl methane sulfonate;  Propane sulfone,  Ethyleneimine  Beta propiolactone  Dimethylsulfate
  • 21. Classical Structural Alerts for Reactive, Electrophilic Direct-Acting Mutagens Cl-CH2-O-CH2-Cl Haloethers Epoxides Strained lactones Sulfonates Enals
  • 22. Mechanisms of Chemical Interaction with DNA: Electrophiles  The majority of genotoxic chemicals that directluy interact with DNA are either electrophiles or are metabolized to electrophiles;  Electrophiles are positively charged species that are attracted to an electron rich center i.e. a nucleophile;
  • 23. Mechanisms of Chemical Interaction with DNA: Electrophiles  An electrophile (literally electron-lover) is a reagent attracted to electrons that participates in a chemical reaction by accepting an electron pair in order to bond to a nucleophile;  Most electrophiles are positively charged, have an atom that carries a partial positive charge, or have an atom that does not have an octet of electrons;
  • 24. Mechanisms of Chemical Interaction with DNA: Electrophiles  Specific areas of DNA behaves like a nucleophile;  DNA nucleophilic centers:  Ring nitrogens of the DNA bases are generally the most reactive centers;  Ring oxygens of the DNA bases are also reactive centers;  S: groups in DNA are also targets  Critically, the N7 of guanine (most reactive); the N3 of adenine and the O6 of guanine are the most common sites
  • 26. Mechanisms of Chemical Interaction with DNA: Electrophiles  Electrophilic damage may also occur to the phosphodiester backbone of DNA:
  • 27. Mechanisms of Chemical Interaction with DNA: Electrophiles  Different electrophiles display different preferences for the various DNA nucleophilic sites and different spectra of damage;  One of the intensively researched concepts is that the DNA damage spectrum for different electrophiles could be used as a marker of environmental exposure: has not worked because of the variable ability of DNA to repair different types of DNA damage plus the often tissue-specific nature of mutation spectra.
  • 28. Mechanisms of Chemical Interaction with DNA: Oxidization  Oxidation of DNA bases is a normal background biological event associated with aerobic metabolism and other cell reactions;  Important endogenous oxidizing agents are: H2O2, superoxide anion, nitric oxide species, lipid peroxides, hydroxyl radicals, Fenton reaction products;
  • 29. Mechanisms of Chemical Interaction with DNA: Oxidization  Endogenous oxidative damage to DNA occurs at a rate of about 120 occurrences per cell per hour in NORMAL cells!  In general, oxidative damage to DNA is repaired with high fidelity at a maximal rate of about 105 base pairs per hour;  Critically, there are a number of antioxidant pathways that limit the amount of endogenously oxidative species that are produced under normal circumstances;
  • 30.
  • 31.
  • 32.
  • 33. Mechanisms of Chemical Interaction with DNA: Oxidization  Oxidative damage to DNA occurs most readily at guanine residues due to the high oxidation potential of this base relative to cytosine, thymine, and adenine;  Most common oxidized DNA adduct is 8- hydroxyguanine followed by thymine glycol.
  • 34.
  • 35. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  Deamination of cytosine:  Spontaneous deamination is the hydrolysis reaction of cytosine into uracil,  Corrected for by the removal of by uracil-DNA glycosylase, generating an abasic (AP) site;  The AP site is then corrected by base excision repair;
  • 36. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  5-methylcytosine: Spontaneous deamination of 5-methylcytosine results in thymine;  This is the most common single nucleotide mutation. In DNA, this reaction can be corrected by the enzyme thymine-DNA glycosylase;
  • 37. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  Guanine: Deamination of guanine results in the formation of xanthine.  Xanthine, in a manner analogous to the enol tautomer of guanine, selectively base pairs with thymine instead of cytosine. This results in a post- replicative transition mutation, where the original G-C base pair transforms into an A-T base pair. Correction of this mutation involves the use of alkyladenine glycosylase during base excision repair;
  • 38. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  Adenine: Deamination of adenine results in the formation of hypoxanthine.  Hypoxanthine, in a manner analogous to the imine tautomer of adenine, selectively base pairs with cytosine instead of thymine. This results in a post-replicative transition mutation, where the original A-T base pair transforms into a G-C base pair;
  • 39. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  Depurination is a chemical reaction of purine deoxyribonucleosides, deoxyadenosine and deoxyguanosine, in which the β-N-glycosidic bond is hydrolytically cleaved releasing a nucleic base, adenine or guanine, respectively.  Deoxyribonucleosides and their derivatives are substantially more prone to depurination than their corresponding ribonucleoside counterparts.  Loss of pyrimidine bases (Cytosine and Thymine) occurs by a similar mechanism, but at a substantially lower rate.
  • 40. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  When depurination occurs with DNA, it leads to the formation of apurinic site and results in an alteration of the structure;  As many as 5,000 purines are lost this way each day in a typical human cell;  [In cells, one of the main causes of depurination is the presence of endogenous metabolites undergoing chemical reactions;
  • 41. Mechanisms of Chemical Interaction with DNA: Hydrolysis - deamination, depurination, and depyrimidination  Apurinic sites in double-stranded DNA are efficiently repaired by portions of the base excision repair (BER) pathway;  Depurinated bases in single-stranded DNA undergoing replication can lead to mutations, because in the absence of information from the complementary strand, BER can add an incorrect base at the apurinic site, resulting in either a transition or transversion mutation;  Depurination is known to play a major role in cancer initiation.
  • 42. Mechanisms of Chemical Interaction with DNA: Intercalation  Intercalation occurs when ligands of an appropriate size and chemical nature fit themselves in between base pairs of DNA;  Intercalating agents are generally polycyclic, aromatic, and planar, and good DNA stains;  Important examples:  Ethidium bromide (DNA stain);  Anticancer agents: proflavine, daunorubicin, doxorubicin, dactinomy cin, thalidomide.
  • 43. Molecular structure of ethidium intercalated between two pairs of adenine- uracil base pairs.
  • 44. Mechanisms of Chemical Interaction with DNA: Intercalation  In order for an intercalator to fit between base pairs, the DNA must dynamically open a space between its base pairs by unwinding;  The amount of unwinding depends on the specific agent;  This unwinding induces local structural changes to the DNA strand resulting in functional changes: inhibition of transcription and replication and DNA repair processes,  Intercalating agents are commonly potent mutagen.
  • 45.
  • 46. Mechanisms of Chemical Interaction with DNA: Intercalating agents  Important examples:  8,9 epoxide of aflatoxin B1;  Acridine dyes.
  • 47. Mechanisms of Chemical Interaction with DNA: DNA cross linking  Crosslinks occur when exogenous or endogenous agents react with two different positions in the DNA;  Crosslinks can occur in the same DNA strand (intrastrand crosslink) or between opposite strands (interstrand crosslink);  Crosslinks between DNA and protein can occur;
  • 48. Mechanisms of Chemical Interaction with DNA: DNA cross linking  Crosslinks impair DNA replication if the crosslink is not repaired;  Mechanisms:  Bifunctional alkylating agents (e.g. methylene dimethanesulphonate, sulphur mustard, methyl methanesulphonate): mostly act on adjacent N7- guanine bases;  Cisplatin: 1,2-intrastrand d(GpG) adducts (via N7- guanine bases);
  • 49. Mechanisms of Chemical Interaction with DNA: DNA cross linking  Mechanisms:  Nitrous acid is formed in the stomach from dietary nitrites (meat prreservatives): forms interstrand DNA crosslinks at the aminogroup of N2 of guanine at CG sequences;  Malondialdehyde from lipid peroxidation: forms etheno adduct-derived interstrand crosslinks;
  • 50. Mechanisms of Chemical Interaction with DNA: DNA cross linking  Mechanisms:  Psoralens: photoactivated by UVA  form covalent adducts with thymine, one type of which is an intrastrand crosslinking reaction targets TA sequences intercalating in DNA and linking one base of the DNA with the one below it. Psoralen adducts cause replication arrest and is used in the treatment of psoriasis and vitiligo;
  • 51. Mechanisms of Chemical Interaction with DNA: DNA cross linking  Mechanisms:  Aldehydes such as acrolein and crotonaldehyde found in tobacco smoke or automotive exhaust can form DNA interstrand crosslinks in DNA.  Formaldehyde (HCHO) induces protein-DNA and protein-protein crosslinks;
  • 52. Mechanisms of Chemical Interaction with DNA: DNA single strand breaks  Single strand breaks are an extremely common phenomenon – thousands of incidents per cell per day;  Mechanisms of production:  Free radical attack (notably with radiation- induced DNA damage);  DNA alykylation (i.e. electrophylic attack);  Many of the mechanisms are poorly understood;
  • 53. Mechanisms of Chemical Interaction with DNA: DNA double strand breaks  Particularly hazardous to the cell because they can lead to genome rearrangements: regarded as the most dangerous of DNA lesions;  Mechanisms are poorly understood, however oxidization and alkylating agents are able to produce these DNA lesions;  DSBs are induced by a number of different mechanisms, including exposure to ionizing radiation, radiomimetic drugs, collapse of replication forks when the replication machinery encounters single-stranded breaks (SSBs) in the template DNA,
  • 54. Micro-DNA Lesions: Small damage with BIG outcomes.
  • 55. The Ability to Chemically Interact With DNA is Not Enough  For a mutation to occur:  Exposure of DNA generally must occur at the right time in the cell cycle;  A DNA lesion must be chemically produced;  The DNA lesion must not be so gross as to prevent DNA replication and/or produce cell death;
  • 56. The Ability to Chemically Interact With DNA is Not Enough  For a mutation to occur:  The lesion must persist in the DNA long enough for at least 1 cell division (i.e. the “fixing” of the mutation within the cell genome; often referred to as the “expression time” in genetox assays);  The DNA lesion must not trigger the G1/S checkpoint (i.e. apoptosis or senescence; more on this in the carcinogenesis sildes);  The DNA lesion must not trigger the intra-S phase checkpoint (if it does, repair is the likely outcome);
  • 57. The Cell Cycle and Mutation: The vulnerability of the S phase.  Small mutations are most likely to occur when the DNA is being copied i.e. S phase;  Reason for this is that the DNA double helix is unwound and the 2 strands are separated  single DNA strands are particularly vulnerable to chemical attack;
  • 58. (A) Nucleoside triphosphates serve as a substrate for DNA polymerase, according to the mechanism shown on the top strand. Each nucleoside triphosphate is made up of three phosphates (represented here by yellow spheres), a deoxyribose sugar (beige rectangle) and one of four bases (differently colored cylinders). The three phosphates are joined to each other by high-energy bonds, and the cleavage of these bonds during the polymerization reaction releases the free energy needed to drive the incorporation of each nucleotide into the growing DNA chain. The reaction shown on the bottom strand, which would cause DNA chain growth in the 3' to 5' chemical direction, does not occur in nature. (B) DNA polymerases catalyse chain growth only in the 5' to 3' chemical direction, but both new daughter strands grow at the fork. The leading strand grows continuously, whereas the lagging strand is synthesized by a DNA polymerase through the backstitching mechanism illustrated. Thus, both strands are produced by DNA synthesis in the 5' to 3' direction.
  • 59. Proteins at the Y-shaped DNA replication fork: These proteins are illustrated schematically in panel a of the figure below, but in reality, the fork is folded in three dimensions, producing a structure resembling that of the diagram in the inset b. Focusing on the schematic illustration in a, two DNA polymerase molecules are active at the fork at any one time. One moves continuously to produce the new daughter DNA molecule on the leading strand, whereas the other produces a long series of short Okazaki DNA fragments on the lagging strand. Both polymerases are anchored to their template by polymerase accessory proteins, in the form of a sliding clamp and a clamp loader. A DNA helicase, powered by ATP hydrolysis, propels itself rapidly along one of the template DNA strands (here the lagging strand), forcing open the DNA helix ahead of the replication fork. The helicase exposes the bases of the DNA helix for the leading-strand polymerase to copy. DNA topoisomerase enzymes facilitate DNA helix unwinding. In addition to the template, DNA polymerases need a pre-existing DNA or RNA chain end (a primer) onto which to add each nucleotide. For this reason, the lagging strand polymerase requires the action of a DNA primase enzyme before it can start each Okazaki fragment. The primase produces a very short RNA molecule (an RNA primer) at the 58 end of each Okazaki fragment onto which the DNA polymerase adds nucleotides. Finally, the single-stranded regions of DNA at the fork are covered by multiple copies of a single-strand DNA-binding protein, which hold the DNA template strands open with their bases exposed. In the folded fork structure shown in the inset, the lagging-strand DNA polymerase remains tied to the leading-strand DNA polymerase. This allows the lagging-strand polymerase to remain at the fork after it finishes the synthesis of each Okazaki fragment. As a result, this polymerase can be used over and over again to synthesize the large number of Okazaki fragments that are needed to produce a new DNA chain on the lagging strand. In addition to the above group of core proteins, other proteins (not shown) are needed for DNA replication. These include a set of initiator proteins to begin each new replication fork at a replication origin, an RNAseH enzyme to remove the RNA primers from the Okazaki fragments, and a DNA ligase to seal the adjacent Okazaki fragments together to form a continuous DNA strand.
  • 60. The Cell Cycle and Mutation: The S phase checkpoint.  The S-phase checkpoint is a surveillance mechanism, mediated by the protein kinases ATR and Chk2 in human cells;  Responds to to DNA damage by co-ordinating a global cellular response necessary to maintain genome integrity;  A key aspect of this response is the stabilization of DNA replication forks, which is critical for cell survival;  A defective checkpoint causes irreversible replication-fork collapse and leads to genomic instability, a hallmark of cancer cells.
  • 61. If DNA replication is blocked (e.g. by a DNA adduct) ssDNA regions at stalled forks continue to grow because MCM (minichromosome maintenance complex) helicase continues DNA unwinding, although uncoupled from DNA synthesis. The ssDNA binds RPA (replication protein A), which triggers the activation of the checkpoint response. This process is initiated by the recruitment of the Mec1/ATR sensor to RPA-coated ssDNA at stalled forks by its regulatory subunit, Ddc2 (ATRIP in human cells). Mec1 then phosphorylates Mrc1 (the homologue of human Claspin), a mediator that transduces the signal from Mec1 to the effector kinase Rad53, which becomes phosphorylated and activated.
  • 62. The Cell Cycle and Mutation: The S phase checkpoints.  The S-phase checkpoint response co-ordinates DNA replication, DNA repair and cell-cycle progression and regulates processes such as firing of replication origins, stabilization of DNA replication forks in response to DNA damage or replicative stress, resumption of stalled DNA replication forks, transcriptional induction of DNA damage response genes, choice of the repair pathway and inhibition of mitosis until replication is completed;  The S-phase checkpoint is required for cellular viability in DNA damage or replicative-stress conditions
  • 63. Types of Mutations: Point mutations.  Point mutation = single base substitution = the replacement of a single base nucleotide with another nucleotide of the genetic material (either DNA or RNA);  Point mutations most commonly occur during S phase (i.e. DNA replication);  The term also includes insertions or deletions of a single base pair which will result in a frame shift mutation;  Classifications:  By type: deletion, transition, insertion, or transversion;  By the effect on function: nonsense, missense and silent;
  • 64. Types of Mutations: Point mutations.  Transversions (beta mutations):  The substitution of a purine for a pyrimidine or vice versa;  Can only be repaired by a spontaneous reversion;  Transitions produce large chemical changes to DNA structure; thus the consequences of this change tend to be more drastic than those of transitions.  Transversions are classically caused by ionizing radiation and alkylating agents.
  • 65.
  • 66. Types of Mutations: Point mutations.  Transitions (alpha mutations):  A point mutation that changes a purine nucleotide to another purine (A ↔ G) or a pyrimidine nucleotide to another pyrimidine (C ↔ T);  Approximately two out of three single nucleotide polymorphisms (SNPs) are transitions;  Transitions can be caused by oxidative deamination and tautomerization;  Although there are twice as many possible transversions, transitions appear more often in genomes, possibly due to the molecular mechanisms that generate them;  5-Methylcytosine is more prone to transition than unmethylated cytosine, due to spontaneous deamination.
  • 67.
  • 68. Types of Mutations: Point mutations.  Nonsense mutations are point mutations in a sequence of DNA that results in a premature stop codon, or a nonsense codon in the transcribed mRNA, and in a truncated, incomplete, and usually nonfunctional protein product;
  • 69. Types of Mutations: Point mutations.  Missense mutations are point mutations in which a single nucleotide is changed, resulting in a codon that codes for a different amino acid i.e. a non-synonymous change (mutations that change an amino acid to a stop codon are considered nonsense mutations, rather than missense mutations). There are 2 possible outcomes:  Conservative mutations: Result in an amino acid change. However, the properties of the amino acid remain the same (e.g., hydrophobic, hydrophilic, etc). At times, a change to one amino acid in the protein is not detrimental to the organism as a whole. Most proteins can withstand one or two point mutations before their functioning changes;  Non-conservative mutations: Result in an amino acid change that has different properties than the wild type. The protein may lose its function, which can result in a disease in the organism.
  • 70. Types of Mutations: Frame shift mutations.  Silent mutations: Code for the same amino acid.  A silent mutation has no effect on the functioning of the protein;  A single nucleotide can change, but the new codon specifies the same amino acid, resulting in an non-mutated protein;  This type of change is also called synonymous change, since the old and new codon code for the same amino acid;  This is possible because 64 codons specify only 20 amino acids;  Different codons can lead to differential protein expression levels.
  • 71.
  • 72. Types of Mutations: Point mutations.  A frame shift mutation (also called a framing error or a reading frame shift) is a genetic mutation caused by indels (insertions or deletions) of a number of nucleotides that is not evenly divisible by three from a DNA sequence;  Remember that it takes 3 DNA and RNA nucleotides to code for a specific amino acid in a protein i.e. the “reading frames” consist of 3 nucleic acid residues;  Thus insertion or deletion of a DNA nucleotide can change the reading frame (i.e. the grouping “3s” of the codons), resulting in a completely different translation from the original;The earlier in the sequence the deletion or insertion occurs, the more altered the protein produced is.
  • 73.
  • 74. Types of Mutations: Point mutations.  Frame shift mutations;  The earlier in the sequence the deletion or insertion occurs, the greater the effect on protein structure. This is because all of the reading frames after the insertion or deletion will be altered i.e. earlier in the relevant DNA coding sequence the error is, the bigger the overall change to the downstream amino acid composition of the protein;
  • 75. Types of Mutations: Point mutations.  Frame shift mutations;  Frameshift mutations will also alter the first stop codon ("UAA", "UGA" or "UAG") encountered in the sequence. The polypeptide being created could be abnormally short or abnormally long, and will most likely not be functional;  Frameshift mutations frequently result in severe genetic diseases
  • 76. DNA Repair Mechanisms.  High level overview only;  The self-repair of DNA is unique amongst biological molecules;  Major types of mammalian DNA repair:  Base excision repair;  Nucleotide excision repair;  Mismatch repair;  Recombinational repair.
  • 77. DNA Repair Mechanisms.  DNA repair capacity varies by mechanism, tissue, organ, individual and species;  Not all DNA adduct types are equal: some are more easily repaired than others and some types cannot be repaired;  Because of the above 2 points, the use of the number of DNA adducts per cell is NOT a reliable predictor of genetic hazard UNLESS there is very detailed information regarding the kinetics of DNA adduct removal in the specific target tissue and target species!
  • 78. DNA Repair Mechanisms.  In general terms, DNA repair are saturatable mechanisms i.e. there is maximal threshold for the amount of DNA that can be repaired within a given set of parameters;  All DNA repair mechanisms have the ability to detect and repair DNA, and if the DNA repair is successful, the impact of the original DNA damage on the animal is reduced or eliminated;
  • 79. DNA Repair Mechanisms:  In general terms, when low levels of DNA damage occur (i.e. below saturation for repair), error-free (high fidelity) repair occurs. When there are large amounts of DNA damage (i.e. above the saturation for repair), error prone DNA repair predominates;  However: there are big species, sex, age, tissue, and organ differences;  The type of DNA adduct has a significant influence over which type of repair predominates;
  • 80. DNA Repair Mechanisms:  Intrinsic variability within human populations  There is a very large variability to repair DNA damage between individuals: up to 65% of average rate in populations without inherited DNA repair defects (e.g. XP);  People with XP have DNA repair capacity of ~1- 2% of normal;  There are differences in DNA repair capacity between rodents and humans: you cannot directly extrapolate results unless you accurate kinetics across the relevant species;
  • 81. DNA Repair Mechanisms: Mechanisms where only one strand is damaged  When only one of the two strands of a double helix has a defect (i.e. only one of the 2 strands has a damaged/missing nucleotide), the other strand can be used as a template to guide the correction of the damaged strand;  These types of DNA repair are called excision repair: remove the damaged nucleotide and replace it with an undamaged nucleotide complementary to that found in the undamaged DNA strand
  • 82. DNA Repair Mechanisms: Base excision repair  Base excision repair:  Repairs damage to a single base caused by oxidation, alkylation, hydrolysis, or deamination;  The damaged base is removed by a DNA glycosylase;  The DNA base is then recognized by an enzyme called AP endonuclease, which cuts the phosphodiester bond;  The missing part is then resynthesized by a DNA polymerase, and a DNA ligase performs the final nick- sealing step;
  • 84.
  • 87. DNA Repair Mechanisms: Base excision repair  Base excision repair:  BER is the major repair pathway involved in the removal of non-bulky damaged nucleotides;  In general BER is a high fidelity process i.e. not error prone;  BER protects both nuclear and mitochondrial DNA;  Heritable defects in BER (particularly DNA polymerase) are associated with cancer.
  • 88. DNA Repair Mechanisms: Nucleotide excision repair  Nucleotide excision repair (NER), which recognizes bulky, helix-distorting lesions;  A specialized form of NER known as transcription- coupled repair deploys NER enzymes to genes that are being actively transcribed;
  • 89.
  • 90. 1) 3 protein complexes are involved in DNA- damage recognition: XPA, XPC-HR23 and RPA. 2) These proteins recruits Transcription factor II (TFIIH) that incorporate two helicases: XPB and XPD that unwinds a 30 bp DNA fragment around the DNA damage. 3) After DNA unwinding, damaged-DNA strand is excised by XPG and the XPF-ERCC1 complex at 3' and 5' sites respectively. 4) After excision, damaged-DNA strand is removed and replaced by re-synthesizing the template complementary DNA strand by polymerase complex (Pol. E/D, replication protein A (RPA) and replication factor C).
  • 91. DNA Repair Mechanisms: Nucleotide excision repair  Mismatch repair (MMR), which corrects errors of DNA replication and recombination that result in mispaired (but undamaged) nucleotides;  MMR functions primarily as a “proof reader” following DNA replication
  • 93.
  • 94. DNA Repair Mechanisms: Nucleotide excision repair  Mismatch repair (MMR), which corrects errors of DNA replication and recombination that result in mispaired (but undamaged) nucleotides;  MMR functions primarily as a “proof reader” following DNA replication
  • 95. Macro DNA Damage:  Macro DNA damage = damage to chromosomes = clastenogenesis e.g. single strand breaks, double strand breaks, sister chromatid exchange, non- homologous end joining, changes in ploidy;  Abnormal chromosome number = aneuploidy;  Increased chromosome number = polyploidy;
  • 96. Macro DNA Damage: Sister chromatid exchange  Exchange of genetic material between two identical sister chromatids or between chromosomes with identical mutations;  Primarily occurs during S phase;  Four to five sister chromatid exchange/chromosome pair/mitosis is within the normal range, 14-100 exchanges is not normal and presents a danger to the organis;  Mediated by the homologous end-joining mechanism fo DNA;
  • 97.
  • 98. Macro DNA Damage: Sister chromatid exchange (equal cross over)  SCEs can be induced by various genotoxic treatments that result in double DNA strand breaks, suggesting that SCEs reflect a DNA repair process i.e. they are measure of DNA damage;  This process is considered to be conservative and error- free, since no information is generally altered during reciprocal interchange by homologous recombination.  Most forms of DNA damage induce chromatid exchange upon replication fork collapse: Holliday model;  Occurs during prophase I of meiosis (pachytene) in a process called synapsis.
  • 99.
  • 100. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  SCE (equal cross over) is the least harmful outcome of a DSB;  Other potentially catestrophic outcomes include due to misrepair of DSBs include:  Inversions;  Interstitial deletions;  Terminal deletions;  Translocations;  Unequal crossovers.
  • 101. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Inversions:  Inversions that involve the centromere are called pericentric inversions;  Those that do not involve the centromere are called paracentric inversions;  Inversions potentially have massive effects on gene function.
  • 102. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Interstitial deletions:  Causes include the following: Losses from translocation; chromosomal crossovers within a chromosomal inversion; unequal crossing over; breaking without rejoining
  • 103. Macro DNA Damage: Other potential outcomes of DNA double strand breaks
  • 104. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Translocation  = a chromosome abnormality caused by rearrangement of parts between nonhomologous chromosomes;  Gene fusion may be created when the translocation joins two otherwise separated genes: very important in carcinogenesis as it may place the coding region of a relatively inactive gene with normally very active gene promoter region  inappropriate upregulation of a gene (alternatively gene silencing may occur);
  • 105. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Translocation:  There are two main types, reciprocal (also known as non- Robertsonian) and non-reciprocal (Robertsonian);  Also, translocations can be balanced (in an even exchange of material with no genetic information extra or missing, and ideally full functionality) or unbalanced (where the exchange of chromosome material is unequal resulting in extra or missing genes).
  • 106. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Reciprocal translocations:  Reciprocal translocations are usually an exchange of material between nonhomologous chromosomes;  Estimates of incidence range from about 1 in 500 human newborns;  Such translocations are usually harmless and may be found through prenatal diagnosis;  However, carriers of balanced reciprocal translocations have increased risks of creating gametes with unbalanced chromosome translocations leading to miscarriages or children with abnormalities.  Most balanced translocation carriers are healthy and do not have any symptoms.
  • 107.
  • 108. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Nonreciprocal (Robertsonian) translocations:  This type of rearrangement involves two acrocentric chromosomes that fuse near the centromere region with loss of the short arms  The resulting karyotype in humans leaves only 45 chromosomes since two chromosomes have fused together
  • 109. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Nonreciprocal (Robertsonian) translocations:  This has no direct effect on the phenotype since the only genes on the short arms of acrocentrics are common to all of them and are present in variable copy number (nucleolar organiser genes). Robertsonian translocations have been seen involving all combinations of acrocentric chromosomes. The most common translocation in humans involves chromosomes 13 and 14 and is seen in about 0.97 / 1000 newborns.  Carriers of Robertsonian translocations are not associated with any phenotypic abnormalities, but there is a risk of unbalanced gametes which lead to miscarriages or abnormal offspring.
  • 110. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Balanced translocations: no genetic material is lost
  • 111. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Unbalanced translocation: genetic material is lost from one chromosome but gained by another. This means that the progeny will either have missing genetic material or extra genetic material
  • 112. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Acentric fragments: a segment of a chromosome that lacks a centromere;
  • 113. Macro DNA Damage: Other potential outcomes of DNA double strand breaks  Acentric fragments:  Because acentric fragments lack a centromere, the cannot attach to the mitotic spindle i.e. acentric fragments are not evenly distributed to the daughter cells in cell division (mitosis and meiosis). As a result one of the daughters will lack the acentric fragment;  Lack of the acentric fragment in one of the daughter cells may have deleterious consequences, depending on the function of the DNA in this region of the chromosome;  In the case of a gamete, it will be fatal if essential DNA is contained in that DNA segment;  In the case of a diploid cell, the daughter cell lacking the acetric fragment will show expression of any recessive genes found in the homologous chromosome.
  • 114. Acentric fragments are lost from the nuclei of cells following mitosis or meiosis. They form a micronucleus.
  • 115.
  • 116. Macro DNA Damage: Changes in chromosome number  Changes to chromosome number can result from:  Nonreciprocal (Robertsonian) translocations;  Errors in chromosomal segregation i.e. a whole chromosome is left behind during anaphase of mitosis or meiosis;  Damage to the mitotic spindle: e.g. griseofulvin, pcalitaxel, colecemid, vinblastine;
  • 117.
  • 118. Macro DNA Damage: Changes in chromosome number  Kinetocore:  The protein structure on chromatids where the spindle fibers attach during cell division to pull sister chromatids apart;  The kinetochore forms in eukaryotes, assembles on the centromere and links the chromosome to microtubule polymers from the mitotic spindle during mitosis and meiosis;  Kinetochores start, control and supervise the striking movements of chromosomes during cell division;
  • 119. Macro DNA Damage: Changes in chromosome number  Kinetocore:  The protein structure on chromatids where the spindle fibers attach during cell division to pull sister chromatids apart;  The kinetochore forms in eukaryotes, assembles on the centromere and links the chromosome to microtubule polymers from the mitotic spindle during mitosis and meiosis;  Kinetochores start, control and supervise the striking movements of chromosomes during cell division;
  • 120. Macro DNA Damage: Changes in chromosome number  Kinetocore:  Kinetochores are critical in initiating/avoiding the spindle checkpoint
  • 121. Macro DNA Damage: Changes in chromosome number  The spindle checkpoint:  The spindle checkpoint (= spindle assembly checkpoint = mitotic checkpoint), is a cellular mechanism responsible for detection of:  Correct assembly of the mitotic spindle  Attachment of all chromosomes to the mitotic spindle in a bipolar manner  Congression of all chromosomes at the metaphase plate.  When just one chromosome (for any reason) remains lagging during congression, the spindle checkpoint machinery generates a delay in cell cycle progression: the cell is arrested, allowing time for repair mechanisms to solve the detected problem. After some time, if the problem has not been solved, the cell will be targeted for apoptosis (programmed cell death), a safety mechanism to avoid the generation of aneuploidy, a situation which generally has dramatic consequences for the organism.
  • 122.
  • 123. Macro DNA Damage: Changes in chromosome number  Micronuclei can also contain whole chromosomes that were improperly attached to the mitotic spindle during anaphase: these are detectable by staining the micronuclei for kinetocores;  There are at least major causes of micronuclei:  The formation of chromosome fragments that lack a centromere (and thus a kinetocore  cannot attach o the mitotic spindle);  The loss of a whole chromosome which has failed to attach to the mitotic spindle or has broken off the mitotic spindle. This effect is produced by spindle agents;  Extrachromosomal double minutes.
  • 124. Macro DNA Damage: Changes in chromosome number  So what the heck is a double minute you ask (no it is not 120 seconds):  Double minutes are small fragments of extrachromosomal DNA, which have been observed in a large number of human tumors;  They are a manifestation of gene amplification during the development of tumors, which give the cells selective advantages for growth and survival;  They frequently harbor amplified oncogenes and genes involved in drug resistance;  Double minutes, like actual chromosomes, are composed of chromatin and replicate in the nucleus of the cell during cell division;  Unlike typical chromosomes, they are composed of circular fragments of DNA, up to only a few million base pairs in size and contain no centromere or telomere.
  • 126. Mechanisms of micronuclei formation. (A) Aneugenic agents prevent the formation of the spindle apparatus during mitosis. The use of these agents generates micronuclei as a consequence of whole chromosomes lagging behind at anaphase. These chromosomes are left out of the cell nucleus at the end of mitosis; The DNA in the micronuclei could balance the nuclear DNA and result in a complete genome, or be additional to the cell's genome. (B) Clastogenic agents induce micronuclei by breaking the double helix of DNA, thereby forming acentric fragments. These fragments are incapable of adhering to the spindle fibres and integrating in the daughter nuclei, and are thus left behind during mitosis. (C) Micronuclei can also contain highly amplified gene sequences, derived from extrachromosomal double minutes (DM) (yellow dots indicate the presence of a DM). (D(I)) Torsion between the two centromeres of a dicentric chromosome would give rise to the formation of an anaphase bridge that is frequently resolved by breakage. The bridge breakage often results in the formation of acentric fragments that are not included in any of the daughter cell nuclei and form one or more micronuclei at the end of mitosis. (D (II)) It has also been described that, instead of breaking, dicentric chromosomes involved in anaphase bridges are sometimes detached from the two centrosomes, left behind at anaphase and sequestered into micronuclei.
  • 127.
  • 128. US EPA Testing Requirements & Tiers Tier Test Types Assessment Function Rapid, low cost screening, Ames (bacterial reverse mutation) typically for agents where In vitro mammalian cell mutation (e.g. human exposure is low. Hazard Identification 1 mouse lymphoma TK) If all results are negative, In vitro chromosome aberration or generally no further micronucleus assay testing is required In vivo testing: at least one or more of: in Required if there is vivo micronucleus, comet assay, in vivo significant human 2 DNA binding, in vivo unscheduled DNA exposure or +ve results in synthesis, transgenic mouse models Tier 1 Required if +ve results in In vivo tests in germ cells (i.e. dominant Tier 2. Provides a basis for 3 lethal, germ cell micronucleus, germ cell hazard assessment of DNA binding, germ cell USD, germ line effects Provides a quantitative Quantitative in vivo tests for germ cell assessment of germ cell mutation (specific locus test, visible or 4 biochemical markers [mouse spot], mutations for use in quantitative risk heritable translocation test in mice) assessments
  • 129. Germ Cell Versus Somatic Mutation: Female germ cell  The timing of exposure is critically important:  Mutation is most likely to occur during the S phase (during DNA replication) i.e. mitosis and meiosis;
  • 130. Germ Cell Versus Somatic Mutation: Female germ cell  The timing of exposure is critically important:  In humans (and most mammals), this means:  During mitosis of oogonium during fetal development (between weeks 4 and 30 in humans; between days 14.5 and 18.5 in the rat and between days 10.5-12.5 in mice);  There are no further S phases until after fertilization and the formation of a zygote;  Remember: DNA repair occurs in oocytes!
  • 131.
  • 132. Germ Cell Versus Somatic Mutation: Female germ cell
  • 133. Germ Cell Versus Somatic Mutation: Female germ cell  Consequences in terms of germ cell mutation:  The critical timing for female germ cell mutation in mammals occurs at the prenatal stage of development!  Ooctyes are RESISTANT to mutation by non- radiomimetic chemicals (i.e. chemicals that do not produce chromosome or chromatid breaks);  Oocytes are susceptible to radiation and radiomimetic chemicals;
  • 134. Germ Cell Versus Somatic Mutation: Male germ cells  Same basic principle holds true: S phase of cell division processes is most susceptible to mutagenesis;  S phase in spermatogenesis occurs during spermatogonial stem cell stage of development;  Unlike in females, spermatogonial stem cell mitosis occurs throughout the life time of males!  Late spermatids and spermatozoa lack DNA repair – also susceptible to unrepaired DNA damage!
  • 135.
  • 136. Germ Cell Versus Somatic Mutation  Overall, males are generally regarded as being at greater risk of generating germ cell mutations than females because of the continuous, life- time replication of spermatogonial stem cells (where as oogonium are only generated in large numbers during prenatal development in females).
  • 137. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Fundamental principles:  These assays are for micro-DNA damage;  Mutant bacterial test strains are created/selected (typically loose the capacity for synthesis of an amino acid) so they require some form of additional supplementation (usually an amino acid) in order to grow – these are called auxotrophs (auxotrophy is the inability of an organism to synthesize a particular organic compound required for its growth);
  • 138. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Fundamental principles:  The mutation required to produce the auxotrophic strain is generally either a point mutation or a frame shift mutation i.e. a small DNA sequence change.
  • 139. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Fundamental principles:  In order for an auxotroph to grow in medium where the supplement is NOT present, they must undergo a reverse mutation in order to regain the capacity to synthesize the essential substance for growth;  Once the bacteria have undergone a reverse mutaiton, they are able to grow in media that DO NOT contain the supplement (typically an amino acid;  These reverse mutations are typically point mutations or frame shifts.
  • 140. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Fundamental principles:  Many test strains have features that make them more sensitive for the detection of mutations:  Specific responsive DNA sequences at the reversion sites;  Increased cell permeability to large molecules  Lack of DNA repair or enhancement of error-prone DNA repair;
  • 141. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Advantages:  Fast;  Cheap;  Quick screening for micro-DNA damage;  Extensive database/library of the effects of a very diverse array of chemicals;  Generally has acceptable false positive/false negative levels (i.e. good sensitivity, specificity, precision and predictive value)  Although many compounds that are positive in this test are mammalian carcinogens, the correlation is not absolute. It is dependent on chemical class and there are carcinogens that are notdetected by this test because they act through other, non-genotoxic mechanisms or mechanismsabsent in bacterial cells.
  • 142. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Although many compounds that are positive in this test are mammalian carcinogens, the correlation is not absolute. It is dependent on chemical class and there are carcinogens that are not detected by this test because they act through other, non-genotoxic mechanisms or mechanismsabsent in bacterial cells.
  • 143. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Disadvantages:  Utilizes prokaryotic cells, which differ from mammalian cells in such factors as uptake, metabolism, chromosome structure and DNA repair processes;  Tests conducted in vitro generally require the use of an exogenous source of metabolic activation. In vitro metabolic activation systems cannot mimic entirely the mammalian in vivo conditions;  The test does not provide direct information on the mutagenic and carcinogenic potency of a substance in mammals.
  • 144. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Limitations:  Difficult to use with substances that are potent bacteriocides or bacteriostats (i.e. substances that block mitosis);  Culture media are hydrophilic i.e. requires specialized techniques for highly lipophilic substances (e.g. petroleum distillates);  Special techniques are required for gases, vapors or substances that evaporate at 37OC;
  • 145. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Metabolic activation systems:  The objective is to replicate at least some of the major biotransformation pathways in vitro;  Classical system is liver S9 fraction:  Rats are dosed with Arochlor 1254 (a PCB mixture that acts as a potent inducer of liver CYP and UGT enzymes via the AhR pathway)  livers are homogenized  centrifuged at 9000 g for 20 minutes  supernatant is collected;  S9 contains cytosol and microsomes (= smooth endoplasmic reticulum): microsomes component contains cytochrome P450 isoforms (phase I metabolism);cytosolic portion contains the major part of the activities of transferases (phase II metabolism)
  • 146. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Metabolic activation systems:  Classical system is liver S9 fraction:  A NADPH-regenerating system or NADPH solution is required to supply the energy demand of the CYP enzymes (powers the CYP cycle);  For the catalytic activity of phase II enzymes, addition of exogenous cofactors is necessary: UDPGA and alamethicin for UGT; acetyl CoA, DTT, and acetyl CoA regenerating g system for NAT; PAPS for ST; and GT for GST;
  • 147. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Bacteria and strains required for OECD 471:  Salmonella typhimurium TA1535 rfa+ uvrB+ hisG46:  S. typhimurium TA1537 rfa+ uvrB+ hisC3076;  S. typhimurium TA98 rfa+ uvrB+ hisD3052 pKM101;  S. typhimurium TA100 rfa+ uvrB+ hisG46 pKM101;  Escherichia coli WP2 trp+ uvrA;  In order to detect cross-linking mutagens it may be preferable to include TA102 or to add a DNA repair-proficient strain of E.coli [e.g. E.coli WP2 or E.coli WP2 (pKM101)]
  • 148. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Bacteria and strains required for OECD 471:So what does this all mean?  His genotype: location of the deletion mutation in the histidine operon (operon is a functioning unit of genomic DNA containing a cluster of genes under the control of a single regulatory signal or promoter. Net result is that the bacterial carrying this mutation cannot synthesize histidine and are auxotrophs);  rfa mutation: A mutation (rfa) in all strains that leads to a defective lipopolysaccharide (LPS) layer that coats the bacterial surface, making the bacteria more permeable to bulky chemicals;  uvrB and uvrA mutations: The uvrB deletion mutation eliminates the accurate excision repair mechanism, thereby allowing more DNA lesions to be repaired by the error-prone DNA repairmechanism. The deletion through the biotin gene makes the bacteria biotin dependent.
  • 149. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Bacteria and strains required for OECD 471:So what does this all mean?  Plasmid pKM101: present in strains TA1535 and TA1538 resulting in the corresponding isogenic strains TA100, TA98, TA97, TA102 and TA104. Plasmid pKM101 enhances chemical and UV-induced mutagenesis via an increase in the recombination DNA repair pathway. The plasmid confers ampicillin resistance, which is a convenient marker to detect the presence of the plasmid;  Insertion of the mutation hisG428 on the multi-copy plasmid pAQl which was introduced in strain TA102 with the aim of amplifying the number of target sites. To enhance the ability of this strain to detect DNA crosslinking agents, the uvrB gene was retained making the bacterium DNA repair proficient
  • 150. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Bacteria and strains required for OECD 471: So what does this all mean?  trp+: Trp operon is that codes for the production of tryptophan. trp+ E. coli are auxotrophs for tryptophan;  Note: all strains except S. typhimurium TA102 are also biotin dependent (i.e. are both histidine and biotin auxotrophs).
  • 151. E. Coli Wp2 uvrA:
  • 152. Notes: • A trace of histidine (or tryptophan depending on the auxotroph) + biotin are incorporated to allow for 1 or 2 rounds of cell replication in order to fix any mutations present; • The plate incorporation method is reputed to increase the assay sensitivity and to allow the testing of suspensions as well as solutions of test article
  • 153. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Modifications to the standard plate incorporation assay:  The preincubation assay: the tester strains are exposed to the chemical for a short time (20 to 30min) in a small volume (0.5ml) of either buffer or S-9 mix, prior to plating on glucose agar minimal medium (GM agar) supplemented with a trace amount of histidine. With few exceptions it is believed that this assay is more sensitive than the plate incorporation assay, because short-lived mutagenic metabolites may have a better chance reacting with the tester strains in the small volume of preincubation mixture, and the effective concentration of S-9 mix in the preincubation volume is higher than that on the plate.
  • 154. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Modifications to the standard plate incorporation assay:  The desiccator assay for liquids and gases: the use of a closed chamber is recommended for testing highly volatile chemicals and gases;  The Kado Salmonella microsuspension assay for testing samples of small volumes;  Testing chemicals in a reduced oxygen atmosphere: anaerobic environments, such as anaerobic chambers, have been used to study mutagenicity of chemicals and fecal samples under reduced oxygen levels.
  • 155. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Modifications to the standard plate incorporation assay:  Fluctuation method: The fluctuation method is performed entirely in liquid culture and is scored by counting the number of wells that turn yellow from purple in a 96-well microplate. If bacteria are able to revert back to metabolic competence they will continue to replicate and turn the liquid media acid. By including a pH indicator in the media, the frequency of mutation is counted as the number of wells out of 96 which have changed color.  The fluctuation method is comparable to the traditional pour plate method in terms of sensitivity and accuracy, however, it does have a number of advantages, namely, allowing for the testing of higher concentrations of sample (up to 75% v/v), increasing the sensitivity and extending its application to low-level environmental mutagens.[20]  The fluctuation method also has a simple colorimetric endpoint; counting the number of positive wells out of a possible 96 wells is much less time consuming than counting individual colonies on an agar plate.
  • 156. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Strain checks:  Histidine dependence (his): streak a loopful of the culture across a GM agar plate supplemented with an excess of biotin. Because all the Salmonella strains are histidine dependent, there should be no growth on the plates.  Biotin dependence (bio): streak a loopful of the culture across a GM agar plate supplemented with an excess of histidine. There should be no growth on the plate except for strain TA102 which is biotin independent.  Biotin and histidine dependence (bio, his): streak a loopful of the culture across a GM agar plate supplemented with an excess of biotin and histidine. Growth should be observed with all strains.
  • 157. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Strain checks:  rfa marker: streak a loopful of the culture across a GM agar plate supplemented with an excess of biotin and histidine. Apply 10 µl of a sterile 0.1% crystal violet solution. All Salmonella strains should show a zone of growth inhibition (crystal violet is a relatively large bacteriocidal molecule [mw = 407.979] which cannot penetrate the bacteria if a normal cell wall is present);  Presence of plasmid pKM101 (ampicilline resistance): apply in the center of a plate 10 µl of ampicilline solution. Streak a loopful of the pKM101-carrying Salmonella culture across an agar plate supplemented with an excess of histidine and biotin. Growth should be observed.
  • 158. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Strain checks:  Spontaneous mutant frequency: use the standard plate incorporation assay procedure without the inclusion of a solvent for determining the spontaneous mutant frequency (negative control) of each of the tester strains. When the spontaneous control values fall outside an acceptable range the genetic integrity of the strain is considered compromised, and a new culture should be isolated.
  • 159.
  • 160. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Controls:  Must have: suitable positive control for non-metabolic activation and metabolic activation + solvent/vehicle negative control;
  • 161.
  • 162. For tests with metabolic activation:
  • 163.
  • 164. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Evaluation of results:  Surviving populations: usually a 2 – 3-log range of doses are used and the highest of these doses is selected to show some degree of bacterial toxicity (background clearing or reduction in the number of spontaneous mutants);  Dose-response phenomena: a mutagen should display a clear dose-related increase in revertant colonies (can be influenced by poor dose range selection);
  • 165. Classical Assays for Genotoxicity: Bacterial reverse mutation assays  Evaluation of results:  Generally speaking a mutagen will produce a positive dose response over at least 3 different concentrations with the hiighest increase in revertants being 2 – 3 times that of the level of spontaneous revertants in the negative control plates;  Pattern: TA-1535 and TA-100 are derived from the same parental strain, thus commonly the responses of these two strains should be nearly identical;  The results (particularly the pattern of reversion) should be repeatable and consistent;  Specific gene sequencing may be of use in some cases.
  • 166. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Basic principle of the assay:  Cells deficient in thymidine kinase (TK) due to the mutation TK+/- → TK-/- are resistant to the cytotoxic effects of the pyrimidine analogue trifluorothymidine (TFT);  TFT is converted to TFT-monophosphate by thymidine kinase  TFT-triphosphate (TFT-TP) is incorporated into DNA, resulting in cytocidal effects.
  • 167. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Basic principle of the assay:  Thymidine kinase proficient cells are sensitive to TFT, which causes the inhibition of cellular metabolism and halts further cell division;  Thus mutant cells are able to proliferate in the presence of TFT, whereas normal cells, which contain thymidine kinase, are not.
  • 168. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Basic principle of the assay:  The TK mutations have no effect on the growth of cells in normal media because normal DNA synthesis does not involve the TK pathway;  The L5178Y cells also have point mutations in both p53 alleles,. Because p53 is an important protein in the DNA damage response in the cell, the L5178Y cell line is inadequate in its response to DNA damage, which is arguably important for enhanced assay sensitivity for the detection of genotoxic compounds.
  • 169. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Basic principle of the assay:  Assay is usually conducted with and without S9 metabolic activation;  Controls:  Vehicle/solvent negative control;  Positive controls: methylmethanesulfonate for studies without metabolic activation; cyclophosphamide, benzo(a)pyrene and 3-methylcholanthrene for tests with metabolic activation;
  • 170. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Dose range:  Selected so that the test doses span the range from 0% to 80% reduction in cell growth with or without S9 activation.
  • 171. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Assay acceptance criteria:  The average absolute cloning efficiency must be in the range of 65 – 120% (ability of single cells to form a new colony);  The average increase in the vehicle control cell population should be 8 -32 fold over 2 days;  Background forward mutation frequency should be acceptable;
  • 172. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Assay acceptance criteria:  The frequency of forward mutation in the positive controls must be consistent with historical data for the lab;  The assay must include applied concentrations that reach 5 mg/mL or 10 mM (which ever is the lower) for test articles that cause little or no cytotoxicity;  The assay must include applied concentrations that reduce the relative cell growth by approximately 20%; OR  Reach a concentration that exceeds the solubility limit.
  • 173. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Interpretation:  Cell colony size on agar: large mutant colonies have few genetic changes other than the tk+/-  tk-/- forward mutation; small mutant colonies generally represent more extensive genetic modification of the cells;  A mutant frequency of at least 2 times that in the negative control is suggestive of a positive control (induction of mutation is an additive process and not a multiple over background – the global evaluation factor should be used);
  • 174. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Interpretation:  Global evaluation factor = the mean of each vehicle control mutant frequency distribution + 1 SD across multiple test labs;  The GEF for the agar assay method = 90;  The GEF for 96 well plate assays = 126;  A positive assay is one where the mutant frequency for the test article equals or exceeds the GEF PLUS there is a statistically demonstrable dose trend present;  A negative assay is one where the mutant frequency for the test article is below the GEF PLUS there is no statistically demonstrable dose trend present
  • 175. Classical Assays for Genotoxicity: Mouse lymphoma L5178Y tk+/- forward mutation assay (OECD 476)  Interpretation:  If only one criterion is met, additional studies are needed to clarify the test outcome
  • 176. Classical Assays for Genotoxicity: Other mammalian cell forward mutation tests(OECD 476)  The HRPT assay:  Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) is a transferase that catalyzes conversion of hypoxanthine to inosine monophosphate and guanine to guanosine monophosphate;  Test cell lines are HRPT+/- i.e. have a single functional HRPT allele that is lost with mutation;  Multiple different mammalian cell lines with this genotype are available: V79 Chinese hamster cells, AS52 Chinese hamster cells, Chinese hamster ovary cells (CHO) and human TK6 human lymphoblastoid cells.
  • 177. Classical Assays for Genotoxicity: Other mammalian cell forward mutation tests(OECD 476)  The HRPT assay:  Basic principles: HRPT converts 6-thioguanine (TG) or 8- azaguanine (AG) to non-toxic metabolites;  Cells that have HRPT function are able to survive and replicate in the presence of TG or AG;  Those cells that lack HRPT function due to a forward mutation die in the presence of TG or AG;  The assay is conducted and assessed in a manner that is similar to the TK assay.
  • 178. Classical Assays for Genotoxicity: Other mammalian cell forward mutation tests(OECD 476)  The XHRPT assay:  Transgene of xanthineguanine phosphoribosyl transferase (XHRPT);  Works on the same principle as the HRPT assay;  XHRPT is located on autosomal chromosomes where as HRPT is located on the X chromosome.
  • 179. Classical Assays for Genotoxicity: Other mammalian cell forward mutation tests(OECD 476)  The TK, HPRT and XPRT mutation tests detect different spectra of genetic events. The autosomal location of TK and XPRT may allow the detection of genetic events (e.g. large deletions) not detected at the HPRT locus on X-chromosomes.
  • 180. Classical Assays for Genotoxicity: In vivo mammalian forward mutation assays in transgenic animals.  The Big Blue mouse assay:  The genome of these mice has been manipulated such that every cell contains, stably integrated into the DNA, multiple tandem copies of a bacterial lac I repressor gene;  lf the mice are exposed to mutagens, there is a small probability that a mutation will occur somewhere along the inserted sequence;  Any mutation will lead to an inactive lac I gene and lac repressor protein, meaning the gene (lacZ) for beta- galactosidase will no longer be repressed;
  • 181. Classical Assays for Genotoxicity: In vivo mammalian forward mutation assays in transgenic animals.  The Big Blue mouse assay:  The DNA is extracted from the tissues of the treated mouse  the vector is isolated and used to make functional bacteriophages  E. coli cells are mixed with the bacteriophage and spread on a solid culture medium  the bacteriophages infect and destroy ("lyze") the E. coli cells --? this causes clear circular zones, called plaques, to appear in a "lawn" of bacteria  Before they die, cells that have been infected by bacteriophages carrying a mutated lac I will produce beta-galactosidase  This reacts with a substrate in the culture medium turning it blue Count both colorless and blue plaques  The number of blue plaques divided by the total number of plaques gives the mutation frequency.
  • 182. Classical Assays for Genotoxicity: In vivo mammalian forward mutation assays in transgenic animals.  The Big Blue mouse assay:  Bacteriophages with non-mutated genes produce colorless plaques because no beta-galactosidase is synthesized;
  • 183.
  • 184.
  • 185. Classical Assays for Genotoxicity: In vivo mammalian forward mutation assays in transgenic animals.  The muta-mouse assay operates in a manner similar to the Big Blue except it uses a LacZ gene
  • 186. Classical Assays for Genotoxicity: In vivo mammalian forward mutation assays in transgenic animals.  In vivo forward mutation assays offer very substantial advantages:  The complete array of metabolic processes are present;  Tissue specific metabolism is taken into account rather than just the over simplified metabolism that occurs with S9;  Tissue and even cell-type specific mutation rates can be measured. This includes germ cell (spermatogonial) mutations in males!;  Takes into account toxicokinetic and toxicodynamic differences between different organs, tissues and cell types;  In general, tissue specific mutation frequencies generally match the distribution of mutation in live animals!
  • 187. Classical Assays for Genotoxicity: In vitro chromosomal aberration assay (OECD 473)  Basic principle of the assay:  Typically Chinese hamster ovary cells strain CHO-WBL ATCC CCL61 are used (fibroblastic cell line);  Chromosomal number of 21 with a low frequency of spontaneous mutations;  Alternatively human peripheral blood lymphocytes that have been stimulated to divide using a mitogen (PHA) are used;
  • 188. Classical Assays for Genotoxicity: In vitro chromosomal aberration assay (OECD 473)  Basic principle of the assay:  Cell cultures are exposed to the test substance both with and without metabolic activation  At predetermined intervals after exposure of cell cultures to the test substance, they are treated with a metaphase- arresting substance (e.g. Colcemid® or colchicine), harvested, stained and metaphase  Cells are analysed microscopically for the presence of chromosome aberrations.
  • 189. Classical Assays for Genotoxicity: In vitro chromosomal aberration assay (OECD 473)  Basic principle of the assay:  Cell cultures are exposed to the test substance both with and without metabolic activation  At predetermined intervals after exposure of cell cultures to the test substance, they are treated with a metaphase- arresting substance (e.g. Colcemid® or colchicine), harvested, stained and metaphase  Cells are analysed microscopically for the presence of chromosome aberrations.
  • 190. Classical Assays for Genotoxicity: In vitro chromosomal aberration assay (OECD 473)  Measurement of results:  This assay does NOT determine aneuploidy (i.e. increased or decreased number of chromosomes; this is because of artifacts associated with the cell analysis preparation): only cells with a normal chromosome number are analysed;  The types of chromosomal aberrations that are detected are:  Simple breaks;  Complex exchanges;  Gaps;  Dicentric chromosomes;  Ring chromosomes.
  • 191. Classical Assays for Genotoxicity: In vitro chromosomal aberration assay (OECD 473)  Advantages:  Accurate identification of all the different chromosome mutation types;  Possible co-detection of mitotic indices;  No full automatic but interactive scoring possible;  Disadvantages:  High false positive rate;  Labor intensive and time consuming;  Heavily dependent on operator/reader skill.
  • 192. Classical Assays for Genotoxicity: In vivo bone marrow chromosomal aberration assay (OECD 475)  Advantages over the in vitro method:  Takes into account in vivo TK and TD to a certain extent;  Can test different routes of exposure;  Full in vivo metabolism (but with some limitations, particularly given that the tissue analyzed is bone marrow);
  • 193. Classical Assays for Genotoxicity: In vivo bone marrow chromosomal aberration assay (OECD 475)  Disadvantages over the in vitro method:  Exposure of the bone marrow must occur in order for the test to be valid – this may need to be demonstrated by a TK study;  If there is evidence that the test substance, or a reactive metabolite, will not reach the targettissue, it is not appropriate to use this test;
  • 194. Classical Assays for Genotoxicity: In vivo bone marrow chromosomal aberration assay (OECD 475)  Disadvantages over the in vitro method:  If metabolism to an ultimate mutagen is required, then the blood and tissue T½ must be long enough for bone marrow exposure to occur unless local metabolism in the bone marrow occurs;  Risk of excessive toxicity producing distorted results: dose ranging studies are often needed.
  • 195. Classical Assays for Genotoxicity: In vivo bone marrow chromosomal aberration assay (OECD 475)  Principle of the assay:  Animals (typically rats or Chinese hamsters) are exposed to the test substance by an appropriate route of exposure and are sacrificed at appropriate times after treatment;  Prior to sacrifice, animals are treated with a metaphase- arresting agent (e.g., colchicine or Colcemid®);  Chromosome preparations are then made from the bone marrow cells and stained, and metaphase cells are analysed for chromosome aberrations.
  • 196. Classical Assays for Genotoxicity: In vivo spermatogonial chromosomal aberration assay (OECD 483)  Assay is similar in principle to OECD 475;  Important difference is that it tests for GERM CELL chromosomal damage;  If there is evidence that the test substance, or a reactive metabolite, will not reach the target tissue, it is not appropriate to use this assay.
  • 197. Classical Assays for Genotoxicity: Unscheduled DNA Synthesis OECD428  This is essentially a test of DNA repair that occurs outside of the normal period of DNA synthesis in cells;  Measures DNA synthesis in cells which are not in the S phase of the cell cycle;  The assay measures global genomic nucleotide excision repair (NER);
  • 198. Classical Assays for Genotoxicity: Unscheduled DNA Synthesis OECD428  The classical OECD assay measures DNA synthesis by measuring the incorporation of 3H-thymidine or BrDU into DNA;  More modern techniques use specific DNA dyes and flow cytometry: faster, more accurate, provides more information;
  • 199. Classical Assays for Genotoxicity: Unscheduled DNA Synthesis OECD428  The test requirements are very broad:  Can be done in vitro or in vivo;  Primary cell cultures or established cell lines can be used;  Test is performed with or without metabolic activation;  In order to discriminate between UDS and normal semi- conservative DNA replication, cell replication is inhibited or minimized using an arginine-deficient medium, low serum content, or by hydroxyurea in the culture medium;  For flow cytometric methods, inhibition of the cell cycle is not required.
  • 200. Classical Assays for Genotoxicity: Unscheduled DNA Synthesis OECD428  Given that the assay measures nucleotide excision repair, it is critical that:  At least some of the DNA lesions produced by the test article are repairable by nucleotide excision repair;  The test cell line/animal line is capable of relatively normal nucleotide excision repair
  • 201. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Basic principle of the assay:  It is an assay of macro-chromosomal damage (chromosome fragments lacking a kinetocore/centromere) OR damage to the mitotic spindle (whole chromosomes chromosome fragments containing a kinetocore);  The assay detects lagging chromosome fragments or lagging chromosomes;
  • 202. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Basic principle of the assay:  Takes advantage of the fact that when bone marrow erythroblast develops into a polychromatic erythrocyte the cell nucleus is extruded from the cell however DNA containing micronuclei are not extruded from the cell;  Micronuclei can be detected by cell staining and flow cytometry. The lack of a normal cell nucleus makes the detection of micronuclei much easier.
  • 203. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  This assay is regarded as the highest tier assay of the commonly conducted genetic toxicology assays for chemicals and other agents;
  • 204. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Important features:  Classically outbred rats and mice are used in preference to inbred strains in order to reduce the likelihood of strain specific responses (classically CD-1 [ICR] BR mice and/or Sprague-Dawley CD [SD] IGS BR rats);  Animals are usually 8 -10 weeks old at the start of the study;  Various routes of administration, including parenteral routes, are available. Unless there are specific reasons not to, the study route of exposure should match those of the likely human routes of exposure;  Parenteral routes (IV, IP) are used if absorption is poor or if there are other technical constraints regarding dosing.
  • 205. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Important features:  The IV route of exposure has a particular advantage in that it guarantees exposure of the bone marrow;  If non-IV routes of exposure are used, there must be sufficient TK data that demonstrates that bone marrow exposure occurs or is likely to occur;  If the test article is a pro-mutagen (i.e. requires metabolic activation), then there must be reasonable certainty that appropriate metabolic activation can occur in the bone marrow OR the active metabolites have a chemical half-life that is long enough for them to reach the bone marrow!
  • 206. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Important features:  Ideally, 2 positive controls should be used:  Cyclophosphamide: requires metabolic activation;  A substance that does not require metabolic activation (e.g. ethyl methanesulfonate)
  • 207. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Positive controls:  Can be administered by a different route than the test article;  Can be administered as a single dose;  Negative controls: typically a solvent or vehicle control
  • 208. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  “No standard treatment schedule (i.e. 1, 2, or more treatments at 24 h intervals) can be recommended. The samples from extended dose regimens are acceptable as long as a positive effect has been demonstrated for this study or, for a negative study, as long as toxicity has been demonstrated or the limit dose has been used, and dosing continued until the time of sampling. Test substances may also be administered as a split dose, i.e., two treatments on the same day separated by no more than a few hours, to facilitate administering a large volume of material.”
  • 209. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  The test can be performed in 2 ways:  “Animals are treated with the test substance once. Samples of bone marrow are taken at least twice, starting not earlier than 24 hours after treatment, but not extending beyond 48 hours after treatment with appropriate interval(s) between samples. The use of sampling times earlier than 24 hours after treatment should be justified. Samples of peripheral blood are taken at least twice, starting not earlier than 36 hours after treatment, with appropriate intervals following the first sample, but not extending beyond 72 hours. When a positive response is recognized at one sampling time, additional sampling is not required.”
  • 210. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  The test can be performed in 2 ways:  “If 2 or more daily treatments are used (e.g. two or more treatments at 24 hour intervals), samples should be collected once between 18 and 24 hours following the final treatment for the bone marrow and once between 36 and 48 hours following the final treatment for the peripheral blood (12).”
  • 211. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Dose rates:  “If a range finding study is performed because there are no suitable data available, it should be performed in the same laboratory, using the same species, strain, sex, and treatment regimen to be used in the main study (13). If there is toxicity, three dose levels are used for the first sampling time. These dose levels should cover a range from the maximum to little or no toxicity. At the later sampling time only the highest dose needs to be used. The highest dose is defined as the dose producing signs of toxicity such that higher dose levels, based on the same dosing regimen, would be expected to produce lethality. “  Toxicity can take any form. A commonly used measure is a reduction in the ratio of polychromatic to normochromatic erythrocytes (what does this say about the bone marrow?);
  • 212. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Dose rates:  “Substances with specific biological activities at low non-toxic doses (such as hormones and mitogens) may be exceptions to the dose- setting criteria and should be evaluated on a case-by-case basis. The highest dose may also be defined as a dose that produces some indication of toxicity of the bone marrow (e.g. a reduction in the proportion of immature erythrocytes among total erythrocytes in the bone marrow or peripheral blood).”  In general, a maximum limit dose of 2 g/kg is used for substances with low toxicity
  • 213. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Sexes:  In general, if there is data that indicates that there are no sex differences in toxicity, only males are used;  If there is data indicating sex differences, BOTH sexes MUST be used.
  • 214. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  Assay acceptance criteria:  % micronucleated PCE in the negative control group must lie within the historical normal range for the lab and must be below 0.4%;  Must be a statistically significant increase in the % micronucleated PCE relative to the negative controls in the positive control group and this increase should be consisted with historical data for the lab;  Either the limit dose is reached or at least some non-leathal toxicity must be present in the highest test article dose
  • 215. Classical Assays for Genotoxicity: Mammalian Erythrocyte Micronucleus Test OECD 474  What constitutes a positive response?  A statistically significant increase in micronucleated PCE in at least one of the test article doses relative to the negative controls; OR  A statistically significant dose response relationship is present;  The results make biological sense.
  • 216. Classical Assays for Genotoxicity: Rare Assays  Mouse Spot Test (OECD 484)  Now extremely difficult to perform because the appropriate mouse strains are no longer available (lost);  It is a high tier assay in terms of risk assessment/hazard classification;  Often now replaced by Big Blue or Mutamouse  An in vivo test in mice in which developing embryos are exposed to a test agent.  The target cells are melanoblasts;  The target genes are those which control the pigmentation of the coat hairs;
  • 217. Classical Assays for Genotoxicity: Rare Assays  Mouse Spot Test (OECD 484)  The developing embryos are heterozygous for a number of these coat colour genes;  A mutation in, or loss of (by a variety of genetic events), the dominant allele of such a gene in a melanoblast results in the expression of the recessive phenotype in its descendant cells, constituting a spot of changed colour in the coat of the resulting mouse.  The number of offspring with these spots, mutations, are scored and their frequency is compared with that among offspring resulting from embryos treated with the solvent only. The mouse spot test detects presumed somatic mutations in foetal cells.
  • 218. Figure 2Defects in melanocyte development cause white spotting, while the stem cell defect results hair graying. (A) A Ednrbs-l/Ednrbs-l mouse demonstrating extensive piebald spotting. (B) A KitW-2J/+ mouse demonstrating a white head blaze, and small dorsal spot on the back. (C) A Sox10Lacz/+ mouse exhibiting the characteristic white belly spot. (D) A Mitfvit/vit mouse (upper) exhibits gradual hair graying. A lower mouse is age- matched control. Adapted from the WEB site of the European Society for Pigment Cell Research (ESPCR), “Color genes”: http://www.espcr.org/micemut/.
  • 219. Classical Assays for Genotoxicity: Rare Assays  Mouse Heritable Translocation Assay (OECD 485)  This is a particularly useful assay because it is an in vivo measure of germ cell macro-chromosomal damage;  Currently there is no commonly used in vivo equivalent to this assay;  There are 2 forms of the assay:  Detection of changes in fertility of the F1 progeny of exposed animals;  Cytogenetic examination of the F1 progeny of exposed animals;
  • 220. Classical Assays for Genotoxicity: Rare Assays  Mouse Heritable Translocation Assay (OECD 485)  The assay primarily measures the frequency of structural chromosome assays that do not result in a loss of genetic material i.e. reciprocal translocations;  Reciprocal translocations = fetal survival + no malformations;  Non-reciprocal translocations usually mean fetal death;
  • 221. Classical Assays for Genotoxicity: Rare Assays  Mouse Heritable Translocation Assay (OECD 485)  Reciprocal translocation, however, result in a loss of fertility in the F1 generation due to the production of unbalanced chromosomes in the gametes (i.e. unbalanced translocations);
  • 222. Classical Assays for Genotoxicity: Rare Assays  Insect and yeast OECD assays: rarely if ever used. You will occasionally come across them in historical data sets. Treat them with caution!
  • 223. Other Common Genotoxicity Assays: The comet assay  = Single Cell Electrophoresis Assay;  Simple, reliable and particularly useful!;  Well accepted assay;  Can also be used as a very simple, but effective and accurate assay of DNA repair;
  • 224. Other Common Genotoxicity Assays: The comet assay  Principle:  Measures DNA strand breaks (i.e. clastenogenesis);  Can be done in vitro or in vivo;  Can be used for specific tissues or even specific cells;  Cells embedded in agarose on a microscope slide are lysed with detergent and high salt to form nucleoids containing supercoiled loops of DNA linked to the nuclear matrix;  Electrophoresis at high pH results in structures resembling comets, observed by fluorescence microscopy;  The intensity of the comet tail relative to the head reflects the number of DNA breaks
  • 225.
  • 226.
  • 227.
  • 228. A Few Slides of Gratuitous Silyness After 228 power point slides on genetic toxicology, I figured you were entitled…..
  • 229.
  • 230.
  • 231.
  • 232. Before Genetic Tox After Genetic Tox
  • 233. ?