alternative methods in pesticides toxicologyrp.ksu.edu.sa/sites/rp.ksu.edu.sa/files/res-files... ·...

49
ALTERNATIVE METHODS IN PESTICIDES TOXICOLOGY Dr. Alaa Eldin Bayoumi Abd-ElKhalek Moustafa

Upload: others

Post on 10-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • ALTERNATIVE METHODS

    IN

    PESTICIDES TOXICOLOGY

    Dr. Alaa Eldin Bayoumi Abd-ElKhalek Moustafa

  • X: Xenobiotic, Pesticide

    EVALUATION OF TOXICITY IN VIVO

    Prediction of the Toxicity on

    the human and the Environment.

    Laboratory Animal and/or

    Aquatic system

    Acute, subchronic, chronic and long-term

    toxicity studies

    High

    Economic

    Coast

    500.000

    to

    1.500.000 $

    Long time

    2-3 years

    1500 Animal

  • mouse 65 %

    rat 22 %

    other rodents 3.19 %

    rabbit 1.62 %

    reptiles and

    amphibians 0.57 %

    fish 4.80 %

    ungulates 2.01 %

    carnivores 0.55 %

    pigs 0.46 %

    monkeys 0.16 %

    DISTRIBUTION OF SUFFERING BETWEEN ANIMAL SPECIES

  • HHISTORY OF THE ALTERNATIVE METHODS IN VITRO

    1798, Jeremy Bentham : The question is not, can they reason, nor can they talk, but can they suffer?

    Century XIX, Marchal Hall: (avoidance the duplication of experiments

    and the inneseccary suffering of animals).

    1959, Three R´s Principles by Russell and Burch: Principles of Human Experimental Technique.

    (Progressive move to reduce, refine and replace the use of animals in experiments)

    1967, UAA (The united action for Animals).

    1969, FRAME (Fund for Replacement of Animals in Medical Experiments).

    1971, European Center of Decumentation and information of the Alternative Methods.

    1986, ECVAM (European Center for Validation of Alternative Methods).

    1995, GTEMA, (Grupo de Trabajo Especializado en Métodos Alternativos).

    Decrease

    of Animals Number

  • PRINCIPLE OF THREE R´s

    William Russell and Rex Burch (1959)

    Progressive move to

    reduce,

    refine and

    replace

    the use of animals in experiments

    Alternatives: methods which reduce the number of animals necessary for an

    experiment, refine existing tests by minimizing animal distress, or replace

    whole-animal use with in vitro other tests.

    3Rs principles

  • Reduction: Cutting the number of animals involved in a scientific experiment to the bare

    minimum that will give results that are

    scientifically valid.

    Refinement: Ensuring that any animals used in experiments experience the minimum of

    suffering, and that their welfare is safe-

    guarded in every possible way.

    Replacement: The use of experimental techniques that do not involve living animals, such as

    human volunteers, cell and tissue

    culture, and computer modelling.

    3Rs PRINCIPLES

  • Any strategy that will result in less animals being used to

    obtain the same amount of information, limiting or avoiding

    the subsequent use of additional animals.

    Appropriate experimental design and appropriate

    analysis of the results based on statistical principles.

    1.- Reduction

  • 1.1.- Environmental enrichment of animal housing.

    1.2.- Laboratory staff must be well trained and competent in the handling

    of the species.

    1.3.- Anaesthesia and analgesia should be used whenever appropriate and

    possible.

    1.4.- Use techniques and parameters which are least invasive and which can

    be assessed as early as possible, in order to minimise both the distress

    caused to the animals and the duration of the study.

    1.5.- At the end of the experiment, the most humane method of euthanasia

    should be chosen.

    2.- Refinement The term refinement signifies the modification of any procedures that

    operate from the time a laboratory animal is born until its death, so

    as to minimise the pain and distress experienced by the animal, and to

    enhance its well-being.

  • Any experimental system which does not entail the

    use of a whole, living animal Is Considered to be a

    replacement alternative.

    3.- Replacement

    3.1.- information

    3.2.- computer-based systems

    3.3.- physico-chemical techniques

    3.4.- the use of lower organisms and embryo stages

    3.5.- cell, tissue and organ cultures

  • SOURCES OF ALTERNATIVE METHODS IN VITRO

    Vertibrates , Animals

    Microorganisms Bacteria : Vibrio fischeri(Microtox ), Pseudomonas, Fungi and Algae. Invertibrates, insects

    *- Cultures of animal embryos

    *- Organs Bath

    *- Organs Perfusion

    *- Explants and Tissue Culture

    *-Cell Cultures Primary Culture or Established Cell lines

    *- Subcellular fractions mitochondria, DNA, enzymes ...etc.

    Embryo Fecunded egg

    Kidney Pancreas

    Liver Brain

    Se

    lecte

    d A

    nim

    al

    Liver

    Data: Information, computer-based systems and physico-chemical techniques

  • CLASSIFICATION OF ALTERNATIVE METHODS

    USING LIVING ORGANISMS

    I- Alternative Methods at Biological Levels: Basal Cytotoxicity Tests: Irritation assay system

    Microtox test system Ames Test Neutral Red Incorporation (NRI) assay Total Cellular Protein content LDH leakage test

    Energy Metabolism Tests: MTT assay Cellular ATP content

    Special Cytotoxicity Tests: Chromosome aberrations Sister Chromatid Exchange Reconstructed Human Skin Mechanism of Cellular Death (apoptosis).

    II- Alternative Methods at Biochemical Levels: Measurement of Lipid peroxidation. Glutathione Redox balance, i.e. cellular glutathione content, (total, reduced or oxidized), cellular glutathione related enzymes (GST, Grd or Gpx).

  • I- Alternative Methods at Biological Levels:

    THE IRRITECTION ASSAY SYSTEM (IAS )

    IAS is a quantitative in vitro test method employed to detect, the ocular and dermal irritation potential of cosmetics, consumer products,

    pharmaceuticals, pesticides and chemical raw materials.

    Irritection data may be obtained in 5 h, vs.

    the 2-3 w required for in vivo studies.

    IAS are highly reproducible, allowing com-

    Parative ranking of samples.

    Studies have demonstrated that IAS results

    are highly correlated with standard Draize

    tests.

  • Vibrio Fischeri, under optimum growth conditions, it is a very

    brightly glowing species.

    THE MICROTOX ASSAY FOR TOXIC ASSESSMENT

    The Microtox toxicity test system exposes natural luminescent bacteria

    (Vibrio fischeri) to increasingly concentrated solutions of test sample. Reductions in the light output of the microorganisms are monitored

    in a temperature controlled photometer, reflecting the toxicity of the sample.

    Test durations range from 5 to 30 minutes permitting a rapid turnaround of

    results. Specialized computer software reduces the data generated to

    estimate a median effective concentration.

    temperature controlled

    photometer

  • The assay is based upon the reversion of mutations in the histidine

    (his) operon in the bacterium Salmonella typhimurium. Strains with mutations in the his operon are unable to grow without added

    histidine. Revertants will grow on minimal medium plates without his.

    This provides a simple, sensitive selection for revertants of his mutants.

    TA1575 is a substitution his mutant

    TA1578 is a frameshift his mutant

    rfa mutation makes the outer membrane more permeable to large

    molecules.

    uvrB mutation eliminates excision repair gene.

    The plasmid pKM101 increases error-prone repair of DNA damage

    The AMES test for genotoxicity

  • The AMES test for genotoxicity (Cont.)

    Plating in His-

    medium

    Growing in His+ liquid medium

    Growth in His- indicates

    genotoxicity

  • SOME SPECIES USED IN PESTICIDE TOXICITY ASSESSMENT

    Some lower species of invertebrates are used as alternatives of re-

    placement in the assessment of environment contamination.

    Daphnia magna

  • CELLULAR TARGETS FOR ASSESSMENT

    Protein synthesis

    Total protein content

    cell viability

    Neutral red uptake (NRU)

    DNA synthesis

    RNA synthesis proliferation

    Mutagenesis

    ATP content

    MTT Energetic metabolism

    ATPase

    Cell membrane

    Leakage (LDH)

  • DIFFERENT IN VITRO CELLULAR MODELS

    Type In vitro/in vivo Type of Standar. Most commonly

    Characteristics culture used cell lines

    Freshly isola- - good - few hours - poor - hepatocytes

    ted cells - karyotipe - suspension - endothelial

    normal - intestinal

    Primary - good - few days, - poor - fibroblasts

    cultures - karyotipe - monolayer - epithelial

    normal

    Finite cell - variable - month/years - good - fibroblasts

    lines - diploid - monolayer - epithelial

    Continuous - poor - years - very - epithelial - -

    monolayer -aneuploid good - transformed

    (established) or tumoral

  • CONTINUOUS CELL LINES USED IN TOXICOLOGY

    Name Origin Characteristics Replication

    CHO Chinese hamster - epithelial-like 15-20 x (7 d)

    Ovary cells - polyploid

    KB Human epidermal - epithelial-like 10-15 x (7 d)

    oral carcinoma - polyploid

    BHK Syrian hamster - fibroblast-like 2 x (4 d)

    kidney - polyploid

    HeLa Human cervix - epithelial-like 15 x (7 d)

    carcinoma - aneuploid

    3T3 Mouse embryos - fibroblastoid 4-6 x (6 d)

    - aneuploid

    V79 Chinese hamster - fibroblastoid 15-20 x (6 d)

    lung - aneuploid

  • Endpoint

    Cell Morphology Cell size and shape

    Cell to cell contacts

    Nuclear size, shape and inclusions

    Nucleolar vacuole formation

    Cytoplasmic vacuole formation

    Cell adhesion Attachment to culture surface

    Dettachment from culture surface

    Cell to cell adhesion

    BIOLOGICAL ENDPOINTS FOR CYTOTOXICITY (1)

  • BIOLOGICAL ENDPOINTS FOR CYTOTOXICITY (2)

    Endpoint

    Cell viability Vital dye uptake

    Trypan blue exclusion

    Cell number

    Replating efficiency

    Cell proliferation Increase in total DNA

    Increase in total RNA

    Increase in total protein

    Colony formation

  • Endpoint

    Membrane damage Loss of enzymes (LDH)

    Loss of co-factors (NADPH)

    Leakage of preloaded cells

    Radioactive precursors Tritium thymidine to DNA

    Tritium uridine to RNA

    Tritium or sulphur amino

    acids to proteins

    BIOLOGICAL ENDPOINTS FOR CYTOTOXICITY (3)

  • The FRAME modified NRI cytotoxicity test

    Balb 3T3-L1 cells from mice

    The basis of this test is that a cytotoxic chemical (regardless of site or

    mechanism of action) will interfere with the cellular lysosomes, hence

    The lysosomes of the affected cells can not accumulate the neutral red

    dye (NR).

    Neutral Red

    dye accumulation

    RN

    lysosomes

    Basal Cytotoxicity Tests:

  • The LDH Leakage test

    u u u u u u u u u

    u u u u

    u u u u u

    u u u u

    u u

    Transfer of

    supernatant

    ELISA-reader

    Incubation

    With the tested

    compound

    (2-48 h)

    Incubation with

    reaction mixture

    Lactate dehydrogenase (LDH) is a

    stable cytoplasmic enzyme present

    in all cells. It is rapidly released in-

    to cell culture when the plasma

    membrane is damaged.

    The use of microplate reader permits the measurement of multiple samples.

    Release

    of LDH

  • RN

    mitochondria

    Reduction by SDH

    Methyl tetrazolium salt

    Formazan

    INTERFERENCE WITH THE METABOLIC PROCESS,

    The basis of this test is that a cytotoxic chemical (regardless of site or

    mechanism of action) will interfere with the cellular mitochondria,

    hence the mitochondria of the affected cells can not transform the

    tetrazolium salt to formazan by its Succinate dehydrogenase (SDH).

    Energy Metabolism Tests: MTT ASSAYS

  • X

    Selection oa animal and tissue Using an established cell line

    (ATCC)

    Cuturing

    Isolation of tissue and cells

    Adding the tested

    compound

    Observation

    Morphological changes Cell counting

  • Sellected Cell line

    Culture and

    Subculture

    Subcultur a 65%

    confluence

    Adding the desired

    Concentration of the

    tested pesticide

    Measurement of the absorbance by ELISA

    0 1 10 100

    4,2

    3,0

    5,0

    6,0

    7,0

    Data analysis

    Sigma

    plot Estimation of toxicity lines

    and calculation of

    cytotoxicity values

    Following the procedure of

    The sellected assay

  • 1 10 100

    3

    4

    5

    6

    7

    Pro

    bit

    LC75 LC50 Comparative Cytotoxicity

    Comparative Biochemistry

    LC25

    LC12,5

    LC6,25

    RELATION BETWEEN

    CYTOTOXIXITY VALUES

    AND BIOCHEMICAL

    PARAMETERS

  • Diclorodiphenylethanes

    HCH derivatives

    Cyclodienes

    Herbicides

    More

    toxic

    Less

    toxic

    PESTICIDE CYTOTOXICITY IN SEVERAL CELL LINES USING

    FOUR ALTERNATIVE METHODS

    Bayoumi A.E. et al. (1999) Rev.Toxicol.

    BF - 2 RTG - 2 CHO - K1

    RN Proteíns RN Proteins RN Proteins MTT ATPase

    TDE TDE TDE TDE TDE TDE TDE TDE

    DDT DDT DDT DDT DDT DDT DDT DDT

    DDE DDE DDE DDE DDE Metoxi. Metoxi DDE

    Metoxi Metoxi Metoxi Metoxi Metoxi DDE DDE Metoxi

    HCH HCH HCH HCH HCH H CH Lind. HCH

    Lind . Lind . Lind . Lind . Lind . Lind . HCH Lind .

    Clord . Clord Endos. Aldrín Clord Clord Aldrín Aldrín

    Endos Hept. Clord Dield. Hept. Hept. Dield. Dield.

    Aldrín Endos Hept Clord. Aldrín Aldrín Endos Clord

    Hept Aldrín Aldrín Hept Dield. E ndos Clord Endos

    Toxaf . Dield. Dield. Endos Endos Toxaf. Hept Hept

    Dield. Toxaf. Toxaf. Toxaf. Toxaf. Dield. Toxaf. Toxaf.

    Atraz. Atraz Atraz Atraz Atraz Atraz Atraz N.D.

    N.D. Paraq. Paraq. Paraq. Paraq. Paraq. Paraq. N.D.

    Compounds

  • Reconstructed skin is a model of human skin, created from human

    cells which are grown in vitro. The culture medium feeds the system and enables the cells to multiply.

    At the final stage of growth, the culture

    is exposed to the open air to foster the

    formation of a corneous layer.

    Epidermis which include melanocytes

    exposed to UV rays actually tan.

    RECONSTRUCTED HUMAN SKIN: AN EXAMPLE OF REPLACEMENT

  • USE OF ARTIFICIAL SKIN FOR TOXICITY ASSAYS

    Reconstructed skin

    contents all cellular

    elements and layers

    appearing in normal

    skin.

  • Cytogenetic test using CHO cultures

    The cytogenetic analysis of in vitro cultured CHO cells can be used as a mutagenicity test for chromosome abberations and Sister Chromatide Exchanges

    (SCE). After completion of the cell cycle stained metaphase preparations are

    examined with the microscope.

    Chormosome Abberations Sister Chromatid

    Exchanges (SCE)

    The sister chromatid exchange

    (SCE) assay is used to evaluate

    whether cytogenetic damage has

    been caused by chemical or

    physical agents. Chemical

    mutagens often induce SCEs at

    concentrations which are lower

    than those required to produce

    significant yields of chromosomal

    aberrations. When cells are

    treated with compounds that

    cause lesions that persist through

    cell division, exchanges will

    occur between the daughter

    chromatids.

    Special Cytotoxicity Tests:

  • MECHANISMS OF CELLUAL DEATH (APOPTOSIS)

    Necrosis

    Normal reversible Swelling

    irreversible

    Swelling Disintegration

    Conserved chromatin

    Mitochondrial changes

    Apoptosis

    Normal Condensation

    (Cell blebbing)

    DNA

    Fragments

    Mitochondrial preserved

    Fragmentation

    Intact membranes

    apoptic bodies

    Formation of

    apoptic bodies

    Nuclear changes

  • Cytotoxic values of chlordane and toxafene that induced

    apoptosis at Levels of NR25 and NR50 in CHO cell line.

    Absorb

    ance (4

    90

    nm

    )

    0,0

    0,1

    0,2

    0,3

    0,4

    0,5

    0,6

    0,7

    0 2 4 6 8 10

    chlordane-NR25

    toxafene-NR25

    A

    0 2 4 6 8 10

    chlordane -NR50

    toxafene-NR50

    B

    *** ***

    *** *** ***

    ***

    **

    *** *** ***

    ***

    **

    ***

    *** ***

    ***

    ***

    **

    ***

    **

    0

    Time (h)

  • Kinetics of p,p´-DDT, endosulfan and dieldrin which induced apoptosis at levels of NR25 (A) and NR50 (B) in CHO-K1 cells.

    Time (h)

    Absorb

    ance (

    49

    0 n

    m)

    0 2 4 6 8 10

    p,p´-DDT endosulfan

    dieldrin

    ***

    ***

    *** ***

    ***

    ** * **

    **

    **

    ** *

    0,0

    0,1

    0,2

    0,3

    0,4

    0,5

    0 2 4 6 8 10

    p,p´-DDT endosulfan

    dieldrin

    **

    **

    **

    ** ***

    ** **

    0,6

    A B

  • APPLICATIONS OF THE ALTERNATIVE METHODS

    Animal Test

    Draize test in

    Rabbit´s eye

    Corrosivity testing

    On rabbit skin

    Neurotoxicity test

    of organopho-

    sphorus

    compounds

    in Chicken

    In vitro Method

    Eye irritation

    Skin corrosion

    Human skin cultures

    NTE esterases

    determination

    in neuroblastoma

    cells

    Degree of Replacement

    Complete replacemnent

    Complete replacement

    Partial replacement for

    Esterase inhibitors

    Regulatory authorities

    acceptance

    EU

    US, Canada

    Worldwide

  • GENERAL SCHEME FOR USING ALTERNATIVE METHODS

    FOR ACUTE TOXICITY RTESTING,

    1- Examination of physicalchemical properties, literature review and QSAR.

    2- Integrate data.

    3- Is information suffiecient for hazard characterization ?

    4- Conduct in vitro biological tests

    5- Integrate all data

    6- Is information suffiecient for hazard characterization ?

    7- In cases where human data are necessary,

    Is information suffiecient to aalow controlled human exposure ?

    8- Conduct animal studies Conduct human trial

    after ethical review

    Integrate all data

    Is information suffiecient for hazard characterization ?

    Yes

    Yes

    No

    No

    No Yes

    No Yes

    Evaluate hazard and use

    Results in safety assessment

    Spielmann

    and Goldberg,

    (1999)

  • II. ALTERNATIVE METHODS AT BIOCHEMICAL LEVELS

    NADPH + H +

    NADP +

    PENTOSES PHOSPHATE CYCLE

    XENOBIOTIC

    MERCAPTURIC ACIDS

    GSSG

    H2O2

    catalase

    O2

    O 2

    - SOD

    O2

    2 GSH

    Gpx

    Gpx: glutathione peroxidase

    Grd: glutathione reductase

    GST: glutathione S-transferase

    SOD: Superoxide dismutase

    Grd

    GST

    H2O

    According to the fact that the majority of pesticides are producing Free

    radicals such as ROS which in turn causing oxidative stress which May lead to

    lipid peroxidation, so that, it is well documented that the glutathione redox

    balance is considered as the most important biomarker That can be used to

    detect the cell or tissue injury caused by free radical.

    Glutathione Redox Balance

  • Glutationasa

    ADP + Pi

    ATP

    Glutamic acid

    -Glutamylcisteina sinthitase

    -Glutamyl cisteine

    Glutathione synthitase

    X

    NADPH + H+

    NADP+

    O2

    H2O

    H2O2 O2

    O2 -

    Superoxid distmutase

    GSSG

    Glutathione Glutathione

    reductase peroxidase

    2 GSH

    Glutathione

    S-transferase

    Catalase

    -GLU-CYS-GLY

    X

    Dipeptidase

    Glutamyltranspeptidase

    Mercapturic acid

    PENTOSES PHOSPHATE CYCLE

  • HCH and LINDANE on Gpx and Grd

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    100

    200

    300

    400

    500

    600

    700

    NR Units

    0 6,25 12,5 25

    Lindano A HCH ***

    ***

    **

    ***

    ***

    Glutathione peroxidase

    NR50 HCH = 55,70 mg/ml NR50 lindane = 144,06 mg/ml

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    0

    40

    80

    120

    160

    NR Units

    0 6,25 12,5 25

    HCH Lindano

    B

    ***

    ***

    **

    Glutathione reductase

    Bayoumi A.E. et al., (2002) Xenobiotica.

  • CYCLODIENE INSECTICIDES ON Gpx AND Grd

    100

    200

    0

    NR Unit

    0 6,25 12,5 25

    chlordane

    heptachlor

    **

    **

    **

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    Glutathione peroxidase

    NR50 chlordane = 22,16 mg/ml NR50 heptachlor = 23,90 mg/ml

    90

    30

    60

    0

    NR Unit

    0 6,25 12,5 25

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    chlordane

    heptachlor

    **

    **

    *

    Glutathione reductase

    Bayoumi A.E. et al., (2001) Comp Biochem Physiol C Toxicol Pharmacol.

  • Glutathione peroxidase

    400

    NR Unit

    0 6,25 12,5 25

    0

    100

    200

    300

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    Aldrín

    Dieldrín

    **

    ***

    **

    ***

    **

    NR50 aldrín = 56,63 mg/ml NR50 dieldrín = 65,96 mg/ml

    (nm

    ole

    s/m

    in/m

    g p

    rote

    inas)

    60

    20

    30

    40

    50

    NR Unit

    0 6,25 12,5 25

    Aldrín

    Dieldrín

    ***

    ***

    **

    *

    Glutathione reductase

    CYCLODIENE INSECTICIDES ON Gpx AND Grd

    Bayoumi A.E. et al., (2002) Comp Biochem Physiol C Toxicol Pharmacol.

  • Pesticides -Glutathione S-transferase

    Ac

    tivit

    y g

    luta

    thio

    ne

    S

    -tra

    ns

    fera

    se

    (nm

    ol/

    min

    /mg

    pro

    tein

    )

    Cytotoxicity (NR25 mg/ml) 10 100 1000

    0

    10

    20

    30

    40

    50

    60

    r =- 0.831

    p,p´-DDT clordano heptachloro

    aldrin

    methoxychlor

    HCH

    endosulfan

    lindane

    atrazina

    paraquat

    p,p´-TDE

  • Cytotoxicity (RN25 mg/ml)

    Activity o

    f glu

    tath

    ione p

    ero

    pxid

    ase

    (nm

    ol/m

    in/m

    g p

    rote

    in)

    10 100 1000

    0

    100

    200

    300

    400

    500

    600

    700

    r = 0,725

    p,p´-TDE

    chlordane

    heptachlore

    p,p´-DDT

    aldrin

    methoxychlore

    HCH

    lindane

    atrazina

    paraquat

    Glutation peroxidasa-Plaguicidas

  • Cytotoxicity (NR25 mg/ml)

    Pesticide- Total Glutathione

    tota

    l G

    luta

    thio

    ne

    (nm

    ol/m

    g p

    rote

    in)

    10

    20

    30

    40

    50

    60

    70

    10 100 1000 0

    p,p´-TDE

    chlordane

    heptachlore

    aldrin

    methoxychlore

    HCH p,p´-DDT

    lindane atrazina

    paraquat r = 0,847

  • ADVANTAGES AND LIMITATIONS OF IN VITRO ALTERNATIVE METHODS

    ADVANTAGES

    Testing is fast and cheap. Controlled testing conditions. Reduction of systemic effects. Reduction of testing in animals. Presenting high data reproducibility. Human cells and tissues can be used. Reduction variability between experiments. Very small amount of test material is required. Utilizing a biological material highly homogenic. Trangenic cells carrying human genes can be used. Time-dependent studies can be performed and samples taken. Permit and facilitate the study of mechanism of action at the cellular level. Same dose range can be tested in a variety of test systems (cells and tissues). LIMITATIONS

    General side and systemic effects (behaviour) cannot be assessed. Chronic effects cannot be tested.

  • EXAMPLES OF PESTICIDES THAT COULD BE

    TESTED USING ALTERNATIVE METHODS

    All the organochlorine compounds and their metabolites, i.e. DDT, TDE, DDE, methoxychlor,

    aldrin, dieldrin, endrin, chlordane, heptachlor, endosulphan,......etc

    All the organophosphorus compounds and their major metabolites, i.e. Malathion, parathion,

    fenthione, diazinon, chlorpyriphos, dichlorvos, methidathion, Acephate, dimethoat, ............etc

    All the carbamate compounds, i.e. methomyl, carbaryl, .........etc

    All the dithiocarbamate compounds which acting as fungicides

    and their metabolites, i.e. maneb, zineb, mancozeb.......etc

    The majority of herbicides, 2,4-D, paraquat, triazines compounds.

    The majority of pyrethroids compounds, i.e. fenvalerate, deltamethrin,

    permethrin, Tetramethrin, .......etc

    Some of antimolting agents, i.e. diflubenzuron, hexaflumurun, pyriproxyfen.

    Some of synthetized insect sex pheromones, i.e. Pectinophora gossypiella and Heliothis armigera