Journal of Clinical Toxicology

Journal of Clinical Toxicology
Open Access

ISSN: 2161-0495

+44 1478 350008

Review Article - (2017) Volume 7, Issue 1

Discovering Capacity in Novel Targets: C. Elegans as a Model Life Form in Toxicological Research

Destefani AC1*, Costa DS2, Zanardo TEC2 and Taufner GH3
1Professor of the Biomedicine Course of the Faculty of Biomedical Sciences of Espírito Santo, Rua Bolivar de Abreu, 48, Campo Grande, Cariacica/ES, Brazil
2Pharmacist, Master student in Biotechnology from the Federal University of Espírito Santo (UFES), Av. Marechal Campos, 1468 – Maruípe, Vitória/ES, Brasil
3Biomedic, Master student in Biotechnology from the Federal University of Espírito Santo (UFES), Av. Marechal Campos, 1468 – Maruípe, Vitória/ES, Brasil
*Corresponding Author: Destefani AC, Department of Biotechnology, Federal University of Espirito Santo (UFES), Av. Marechal Campos, 1468-Maruipe, Vitória, ES, Brazil, Tel: +55027999434083 Email:

Abstract

The nematode Caenorhabditis elegans has risen as a critical creature show in different fields including neurobiology, formative science, and hereditary qualities. Qualities of this creature demonstrate that have added to its prosperity incorporate its hereditary manipulability, invariant and completely depicted formative program, all around portrayed genome, simplicity of support, short and productive life cycle, and little body estimate. These same elements have prompted to an expanding utilization of C. elegans in toxicology, both for robotic reviews and highthroughput screening approaches. We depict a portion of the exploration that has been completed in the territories of neurotoxicology, hereditary toxicology, and ecological toxicology, and additionally high-throughput tries different things with C. elegans including all inclusive screening for sub-atomic focuses of harmfulness and fast danger evaluation for new chemicals. We contend for an expanded part for C. elegans in supplementing other model frameworks in toxicological research.

Keywords: Caenorhabditis elegans; Neurotoxicity; Genotoxicity; Environmental toxicology; High-throughput methods

Introduction

Caenorhabditis elegans is a saprophytic nematode animal types that has frequently been portrayed as possessing soil and leaf-litter situations in many parts of the world [1-9]; late reports show that it is regularly conveyed by earthly gastropods and other little creatures in the dirt environment [2,3,10,11]. Albeit logical reports on the species have showed up in the writing for over 100 years [4,12], the distribution of Brenner's original hereditary qualities paper [8,13] flagged its rise as a critical test display. Work with C. elegans has since driven in a brief span traverse to fundamental revelations in neuroscience, advancement, flag transduction, cell passing, maturing, and RNA obstruction [5,14]. The accomplishment of C. elegans as a model has pulled in expanded consideration also in the fields of in biomedical and ecological toxicology.

Plainly, C. elegans will be a significant harmfulness demonstrates just if its outcomes were prescient of results in higher eukaryotes. There is expanding proof this is the situation both at the level of hereditary and physiological likeness and at the level of genuine danger information. A number of the fundamental physiological procedures and stress reactions that are seen in higher living beings (e.g., people) are saved in C. elegans . Contingent upon the bioinformatics approach utilized, C. elegans homologues have been recognized for 60–80% of human qualities [15,6], and 12 out of 17 known flag transduction pathways are rationed in C. elegans and human (NRC, 2000). We talk about particular cases in the ranges of neurotoxicology and hereditary toxicology in this audit.

Pathways

The pathways involved in early development are Wnt pathway via β- catenin; Receptor serine/threonine kinase (tumor growth factor-β receptor) pathway; Receptor tyrosine kinase pathway (small G-protein [Ras] linked); Notch-delta pathway and Receptor-linked cytoplasmic tyrosine kinase (cytokine) pathway. Another hand the pathways involved in later development (e.g., organogenesis and tissue renewal) is Apoptosis pathway (cell death pathway) and Receptor protein tyrosine phosphatase pathway. Pathways involved in the physiological function of differentiated cells of the fetus, juvenile, and adult include G-protein–coupled receptor (large G-protein) pathway; Integrin pathway; Cadherin pathway; Gap junction pathway and Ligand-gated cation channel pathway. Signal transduction pathways that are not conserved in nematodes and vertebrates (Table 1) include the Wnt pathway via c-Jun N-terminal kinase, the Hedgehog pathway (patched receptor protein), the nuclear factor kappa-B pathway, the nuclear hormone receptor pathway, the receptor guanylate cyclase pathway, and the nitric oxide receptor pathway (NRC 2000).

Pathways involved in early development
Wnt pathway via β-catenin
Receptor serine/threonine kinase (tumor growth factor-β receptor) pathway
Receptor tyrosine kinase pathway (small G-protein [Ras] linked)
Notch-delta pathway
Receptor-linked cytoplasmic tyrosine kinase (cytokine) pathway
Pathways involved in later development (e.g., organogenesis and tissue renewal)
Apoptosis pathway (cell death pathway)
Receptor protein tyrosine phosphatase pathway
Pathways involved in the physiological function of differentiated cells of the fetus, juvenile, and adult
G-protein–coupled receptor (large G-protein) pathway
Integrin pathway
Cadherin pathway
Gap junction pathway
Ligand-gated cation channel pathway

Table 1: Signal Transduction Pathways Conserved in Nematodes and Vertebratesab. aAdapted from NRC (2000). bSignal transduction pathways that are not conserved in nematodes and vertebrates include the Wnt pathway via c-Jun N-terminal kinase, the Hedgehog pathway (patched receptor protein), the nuclear factor kappa-B pathway, the nuclear hormone receptor pathway, the receptor guanylate cyclase pathway, and the nitric oxide receptor pathway.

Caenorhabditis elegans has various components that make it not simply significant but rather very effective as a model for natural research. Most importantly, C. elegans is simple and reasonable to keep up in research facility conditions with an eating routine of Escherichia coli. The short, bisexual life cycle (3 days) and extensive number (300+) of posterity of C. elegans permits expansive scale generation of creatures inside a brief timeframe [9,1]. Since C. elegans has a little body estimate, in vivo tests can be led in a 96-well microplate. The straightforward body additionally permits clear perception of all cells in develop and creating creatures. Besides, the seriously contemplated genome, finish cell heredity outline, (KO) mutant libraries, and built up hereditary philosophies including mutagenesis, transgenesis, and RNA obstruction (RNAi) give an assortment of choices to control and study C. elegans at the sub-atomic level [14,5].

Since invert hereditary and transgenic tests are much less demanding and less costly to lead in C. elegans when contrasted with numerous other model frameworks, it is a valuable model for atomic examinations of the reaction of moderated pathways to in vivo compound introduction. As an in vivo demonstrate, C. elegans gives a few attributes that supplement in vitro or cell models. The utilization of entire life form measures, as a matter of first importance, permits the investigation of a practical multicellular unit, for example, a serotonergic neurotransmitter, rather than a solitary cell [15,6]. Caenorhabditis elegans additionally empowers the recognition of living being level end focuses (e.g., nourishing, proliferation, life traverse, and motion) and the communication of a concoction with numerous objectives in a living being. In this manner, C. elegans supplements both in vitro and in vivo mammalian models in toxicology.

Of note, these qualities encourage high-throughput explores that can look at both major danger, which are basic since such a large number of chemicals still can't seem to be completely tried, and the quality and quality environment communications whose significance is recently starting to be acknowledged in toxicology.

Here we survey three noteworthy uses of C. elegans in biomedical and ecological toxicology: (1) unthinking toxicology, with an emphasis on neurotoxicity and genotoxicity; (2) high-throughput screening abilities; and (3) natural toxicology and ecological evaluation. We accentuate investigations of neurotoxicity since they are the range of toxicology in which C. elegans has been most abused to date.

Caenorhabditis elegans is well suited for neurophysiology analysis of neurotoxicity. With 302 neurons speaking to 118 portrayed neuronal subtypes [16,7], C. elegans gives an in vivo model to examining components of neuronal damage with determination of single neurons. It initially experienced broad improvement as a model life form keeping in mind the end goal to concentrate the sensory system [13,8], and its neuronal ancestry and the entire wiring graph of its sensory system are stereotyped and completely portrayed [17,18]. Every neuron has been relegated a code name comparing to its area. For instance, ADEL portrays the dopaminergic (DAergic) head neuron "foremost deirid left." This generally "basic" sensory system is included 6393 compound neurotransmitters, 890 electrical intersections, and 1410 neuromuscular intersections [19]. Moreover, the principle neurotransmitter frameworks (cholinergic, γ-aminobutyric corrosive (GABA)ergic, glutamatergic, DAergic, and serotoninergic) and their hereditary systems (from neurotransmitter digestion system to vesicle cycling and synaptic transmission) are phylogenetically rationed from nematodes to vertebrates, which takes into consideration discoveries from C. elegans to be extrapolated and further affirmed in vertebrate frameworks.

A few qualities required in neurotransmission were initially recognized in C. elegans . This is exemplified by the GABA vesicular transporter unc-47 and the administrative translation consider unc-30 (for audit on the GABAergic framework) [20], the vesicular acetylcholine (ACh) transporter unc-17 (for survey on the cholinergic framework) [21], the glutamate-gated chloride channel subunits α1 and β (glc-1 and glc-2, separately, for survey on the glutamatergic framework) [22], and the synaptic proteins unc-18, unc-13, unc-26 (for audit on synaptic capacity) [23]. Tests testing the C. elegans sensory system by laser removal of individual neurons/axons, introduction to drugs, and other outside boosts have encouraged the outline of vigorous behavioral tests to survey the capacity of characterized neuronal populaces [21,24-29]. For instance, inhibitory GABAergic and excitatory cholinergic engine capacities are evaluated by measuring the sinusoidal development (plentifulness and recurrence of body curves) and searching conduct of the worm. Engine and mechanosensory elements of glutamatergic neurons are assessed by measuring the pharyngeal pumping rate and the reaction to touch. Mechanosensory elements of DAergic and serotoninergic neurons are assessed by watching the capacity of worms to back off when they experience nourishment. Moreover, the production of transgenic strains communicating fluorescent proteins in characterized neurons permits in vivo imaging of any craved neuron. While tentatively difficult in the cells of tiny creatures, electrophysiology studies can be directed without breaking a sweat and accomplishment in live worms and refined C. elegans neurons, building up that they are electrophysiologically practically identical to vertebrate neurons in their reaction to different medications [30-32,22]. Given the relative simplicity with which quality KO and transgenic creatures can be produced, the capacity to culture embryonic or essential C. elegans cells offers remarkable viewpoints for neurotoxicology applications and study plans.

The protection of neurophysiologic segments from nematodes to people generally depends on shared hereditary systems and formative projects. Consequently, the accessibility of mutants for a large number of the C. elegans qualities encouraged huge advance in unwinding of developmentally saved cell and hereditary pathways in charge of neuron destiny specificity [16,7], separation [33], movement [34], axon direction [35,36], and synaptogenesis [37,38]. As of late, laser axotomy in C. elegans has been effectively connected to distinguish axon recovery components [39,40], which are of most extreme significance in creating medications to invert neurodegenerative procedures and spinal rope wounds. Basic cell capacities applicable to neurotoxicity studies are likewise monitored. This is best exemplified by the unthinking explanation of the apoptotic pathway in C. elegans , for which the 2002 Nobel Prize in Physiology or Medicine was granted [41-43]. The pathway is of direct enthusiasm to neurotoxicologists since apoptosis is embroiled in numerous neurodegenerative maladies and toxicant-initiated cell death [44-49]. Pathways pertinent to oxidative stress–related neuronal wounds, for example, the p38 mitogen-initiated protein kinase and AKT flagging falls, the ubiquitinproteasome pathway, and the oxidative anxiety reaction are likewise rationed in the worm [50-59].

The nematode model is additionally managable to fascinating hereditary adjustments. Henceforth, it is anything but difficult to produce transgenic worms communicating any sort of mutant recombinant protein, giving intends to the investigation of neurodegenerative infections (see extra examination underneath). Quality KO and modified capacity transformations are as a rule accessible from the Gene Knockout Consortium or the National BioResource Project of Japan (as of now 1/3 of the 20,000 add up to qualities in C. elegans ) [14,4] or on the other hand are advantageously created utilizing chemicals, radiations, or transposons (examined underneath under Caenorhabditis elegans and Genotoxicity). Thus, traditional ways to deal with clarify intracellular pathways in C. elegans incorporate forward and modifier screens taking after arbitrary mutagenesis [60-64]. At long last C. elegans is managable to sexual orientation control (conceivable era of guys, feminized guys, masculinized bisexuals, or feminized bisexuals) allowing thinks about on sex specificity systems of neurotoxicants or scatters and "revival" by driving improvement through the peaceful dauer larval stage [65].

A long time before the most recent technologic advancements (RNAi and high-throughput procedures), C. elegans was utilized to study harmfulness instruments of ecological components influencing the sensory system. The accompanying area gives a summary of the accessible writing on neurotoxicity-related issues tended to in C. elegans . It is not intended to be comprehensive but instead to represent average reviews that are agreeable in the C. elegans stage. We highlight examines with presentation results to different metals and pesticides, and in addition general contemplations on investigations of neurodegenerative sicknesses. We underscore the utility of C. elegans in tending to speculation driven components of neurotoxicity and extrapolations to vertebrate frameworks.

Caenorhabditis elegans has been utilized as a model framework to explain the danger and toxicological instruments of different overwhelming metals, for example, Aluminum (Al), Arsenic (As), Barium (Ba), Cadmium (Cd), Copper (Cu), Lead (Pb), Mercury (Hg), Uranium (U), and Zinc (Zn). By and large, these reviews concentrated on different harmful end focuses, for example, lethality, propagation, life traverse, and protein expression. Some concentration has additionally been coordinated to the impacts of these metals on the sensory system by surveying conduct, journalist expression and neuronal morphology. We give here a couple of cases of these methodologies. Examiners have played out various reviews to survey conduct instigated adjustments taking after presentation of the worm to substantial metals. Contingent upon the end point evaluated, neurotoxic impacts on particular neuronal hardware can be gathered.

For example, an imperfection in headway mirrors an impedance of the neuronal system framed by the interneurons AVA, AVB, AVD, and PVC giving contribution to the An and B-sort engine neurons (in charge of forward and in reverse development) and the inhibitory Dsort engine neurons required in the coordination of development Riddle et al. By recording short recordings and in this way dissecting them utilizing PC following programming, it has been conceivable to measure the general development of C. elegans (remove voyaged, directional change, and so on.), body twists and head whips, upon metal medications, permitting to further correspond the information with harms to neuron hardware. These PC following reviews demonstrated that worms showed a measurement subordinate lessening in locomotory development upon presentation to Pb [66-68] and Al [68], while an expansion in velocity was seen upon introduction to low groupings of Hg as contrasted and Cu [69]. Another review demonstrated that presentation to Ba disabled both body curve and head whipping rates in a measurements subordinate way [70], substantiating mammalian information on the impact of Ba on the sensory system ascribed to its capacity to square potassium channels [71].

Bolstering conduct has likewise been appeared to be influenced upon substantial metal introduction. Sustaining requires an alternate neuronal hardware including M3 (required in pharyngeal unwinding), MC (control of pumping rate), M4 (control of isthmus peristalsis), NSM (invigorate nourishing), RIP, and I neurons Riddle et al. An abatement in sustaining was watched when worms were presented to Cd or Hg [72-75].

Behavioral research concentrate the impact of substantial metals on C. elegans has additionally taken the course of surveying the capacity of the worm to detect the poison and modify its conduct as needs be, including other neural hardware, for example, the amphid and phasmid neurons in charge of chemosensation [76]. By producing fixation gradient–containing plates, Sambongi et al. found that C. elegans could maintain a strategic distance from Cd and Cu however not Ni and that the amphid ADL, ASE, and ASH neurons were in charge of this shirking as their joined removal disposed of the evasion phenotype. Promoting the examination concerning the part of ASH neurons, analysts found that a calcium (Ca2+) convergence could be evoked after uncovering the C. elegans to Cu, which may give understanding into the instrument of the capacity of the worm to show shirking practices [77].

Caenorhabditis elegans shows both here and now and long haul learning-related practices in light of particular tangible sources of info [78], which include characterized neuronal systems. For instance, thermosensation-related learning and memory depend on the AFD tactile neuron sending contributions to the AIY and AIZ interneurons, whose signs are coordinated by the RIA and RIB interneurons to charge the RIM engine neuron [79]. While surveying the capacity of this hardware, worms developed and sustained at an unequivocal temperature are moved to a nourishment denied test plate presented to a temperature angle. The capacity of the worms to discover and stay in the territory of the test plate relating to the nourishing temperature mirrors the working of the thermosensation learning and memory arrange previously mentioned [79]. Strikingly, worms presented to Al and Pb display poor scores at this test, characteristic of a huge lessening of the worms' learning capacity [80]. This restates the learning deficiencies saw in youthful patients overexposed to similar metals [81,82].

While behavioural testing was educational of the neuronal hardware influenced by overwhelming metals, extra analyses revealed the subatomic components of their neurotoxic impacts. For instance, in the beforehand depicted review, in the wake of discovering that Al and Pb prompted memory shortfalls, the agents demonstrated that the cell reinforcement vitamin E successfully turned around these deficiencies, showing a part of oxidative worry in Al and Pb neurotoxicity [80]. The contribution of oxidative worry in metal-incited lethality was further affirmed when worms changed in glutamylcysteine synthetase (gcs-1), the rate-constraining protein in glutathione amalgamation showed extreme touchiness to as introduction when contrasted with wild-sort creatures [83].

Contemplates directed in mammalian models found that Hg can piece Ca2+ channels. In neurons, this blockage can prompt unconstrained arrival of neurotransmitters [84]. In C. elegans , the Ca2+ channel blocker verapamil was found to secure against Hg introduction, proposing that Ca2+ flagging assumes a part in the harmfulness of Hg in this model creature as in warm blooded creatures [85].

Perception of neuron morphology taking after overwhelming metal introduction was additionally performed utilizing C. elegans strains communicating the green fluorescent protein (GFP) in discrete neuronal populaces. Tests utilizing drained U evoked no changes in the DAergic sensory system of C. elegans , a perception certified with information from mammalian essential neuronal societies [86,87]. Then, kel-8 and numr-1, which are included in imperviousness to Cd harmfulness, were upregulated upon Cd presentation. Specifically, GFP levels of KEL-8::GFP and NUMR-1::GFP were expanded in the pharynx and the digestive system notwithstanding the constitutive expression saw in AWA neurons [88-91]. Besides, numr-1 was appeared to be instigated in light of substantial metals, for example, Cd, Cu, Cobalt (Co), Chromium (Cr), Ni, As, Zn, and Hg. NUMR-1::GFP was restricted to cores inside the digestive tract and the pharynx and colocalized with the anxiety responsive warmth stun translation calculate HSF-1::mCherry (Tvermoes and Freedman). This demonstrates these specific qualities were adjusted in light of substantial metals and this may help in the comprehension of the poisonous quality of or the security against these specialists.

Right now, there are over a hundred sorts of pesticides accessible and generous endeavours have been advanced to analyse the neurotoxicity of these specialists. Similitude in neural hardware and the preservation in hereditary cosmetics between C. elegans and people have prompted to various late reviews on pesticide neurotoxicity in this species. In this area, we examine the impacts of three gatherings of pesticides on neurological pathways in C. elegans and their importance to comprehension systems of human neurotoxicity.

Paraquat, otherwise called methyl viologen (mev), is chiefly utilized as a herbicide. Expanded attentiveness toward the potential human dangers related with paraquat presentation comes from studies demonstrating that subjects encountering introduction to this and different herbicides/bug sprays have a higher pervasiveness of Parkinson malady (PD) [92] And expanded mortality from PD [93]. The utilization of C. elegans to concentrate the etiology of PD will be examined in the later area. This is because of the specificity with which these pesticides focus on the nigrostriatal DAergic framework through a height of dopamine and amine turnover [94]. All types of paraquat are effortlessly lessened to a radical particle, which produces superoxide radical that responds with unsaturated layer lipids (Uversky), a presumable instrument of neurotoxicity. Caenorhabditis elegans , has an all-around characterized, yet straightforward DAergic arrange, comprising of eight neurons in the bisexual and an extra six neurons situated in the tail of the male [27] and four DA receptors. Dopamine is known to be required in the tweak of velocity and in learning in C. elegans [95-97]. To date, a few paraquat/mev–altered strains have been produced to study potential pathways in which paraquat applies its poisonous impacts. mev-1 (changed for the succinate dehydrogenase) [98,99] and mev-3 [100] were created to begin with, and both strains showed expanded affectability to paraquat-and oxygen-interceded harm therefore of expanded generation of superoxide radicals [101,102] and extreme touchiness to oxidative anxiety. mev-4 [103], mev-5, mev-6, and mev-7 [104] showed imperviousness to paraquat. Be that as it may, since the proteins that are encoded by these qualities are right now obscure, future mapping of these qualities will probably uncover pathways included in paraquat lethality.

Paraquat applies oxidative harm in vertebrates, which has likewise been confirmed in C. elegans . Mutants that need cell reinforcement proteins, for example, cytosolic or mitochondrial superoxide dismutases (turf 1 and grass 2) demonstrate expanded affectability to paraquat [105], though mutants with expanded superoxide dismutase levels, for example, age-1 (encoding the synergist subunit of phosphoinositide 3-kinase) [106,107] and worms overexpressing the omega-class glutathione transferase gsto-1 [108] show expanded imperviousness to paraquat poisonous quality. In addition, C. elegans mutants excessively touchy to oxygen lethality, for example, rad-8 [109,110] or those with a drawn out life expectancy, for example, daf-2 (encoding insulin/insulin development calculate receptor) [111,112] demonstrate expanded resilience to paraquat. Taken together, these outcomes give novel data on components by which paraquat intercedes its poisonous quality (by upgrading affectability to oxygen harmfulness with a height underway of receptive oxygen species and shortening life expectancy) and give headings to future examinations on systems that prompt to DAergic neurodegeneration.

A moment omnipresent pesticide is rotenone; it is a normally happening and biodegradable pesticide successful in slaughtering irritations and fish [113] f. Specialists initially announced in 2000 that Iv introduction to rotenone may lead in people to the improvement of PD-like side effects joined by the specific annihilation of nigral DAergic neurons [114]. Since rotenone acts by hindering mitochondrial NADH dehydrogenase inside complex I [115], the improvement of a mutant C. elegans strain that shows mitochondrial restraint gave an exploratory stage where the part of this compound could be specifically assessed. A transformation in a 49-kDa subunit of mitochondrial complex I in C. elegans mutant gas-1 shows touchiness to rotenone and oxygen [116], highlighting the significance of a useful complex I in rotenone resistance. Also, C. elegans with adjustments in PD causative qualities are very delicate to rotenone harmfulness, recommending the capacity of these proteins to secure against rotenone-initiated oxidative harm in DAergic neurons [117] (see neurodegenerative sickness segment underneath).

The organophosphates (OPs) are a gathering of bug sprays that objective the cholinergic framework. ACh is the essential neurotransmitter required in engine work in many creatures, including the nematode (Rand and Nonet,). Because of the inclusion of the neuromuscular framework, a PC following framework was utilized to concentrate the neurobehavioral changes in C. elegans related with two OP pesticides (malathion and vapona). Caenorhabditis elegans demonstrated a surprising decrease in velocity at a fixation beneath survival lessening [118,119]. Correlation ponders utilizing comparative behavioral examinations were later created to survey development modification as a marker of the neurotoxity of 15 OP pesticides [120] and carbamate pesticides, which dissimilar to OP pesticides are reversible AChE inhibitors [121]. The LD50 values in C. elegans firmly related with LD50 in both rats and mice. Pesticides (vapon, parathion, methyl parathion, methidathion, and funsulfothion) that indicated cholinesterase hindrance were related with articulated behavioral poisonous quality (i.e., diminish in development). A current review has looked at end focuses utilizing OPs and observed AChE restraint to be the touchiest pointer of poisonous quality additionally the most hard to gauge [122]. Decrease in development for 10 OPs was found to relate to rodent and mouse intense lethality information. At long last, reproduction thinks about inspecting the rate of retention and biodegradation of OP (parathion) additionally [123] build up the importance and unwavering quality of C. elegans as a trial model and indicator for soil lethality.

As already expressed, the C. elegans sensory system practically reiterates large portions of the qualities of the vertebrate cerebrum. Specifically, it can experience degeneration through saved components and is in this manner an effective model for revealing the hereditary premise of neurodegenerative issue. In this area, we will concentrate on PD, Alzheimer disease (AD), Huntington disease (HD), and Duchenne strong dystrophy (DMD).

PD is a dynamic, neurodegenerative turmoil harrowing 2% of the U.S. populace [124]. Trademark highlights incorporate a progressive loss of engine capacity because of the degeneration of DAergic neurons inside the substantia nigra standards compacta and loss of DAergic terminals in the striatum [125]. At the cell level, affidavit of cytoplasmic Lewy bodies made out of collected protein, for example, α-synuclein, is watched. PD cases are alluded as familial (FPD) or idiopathic (IPD) contingent upon whether the ailment is innate (FPD) or from obscure source, perhaps because of natural presentation to neurotoxicants (IPD) [126,127]. Among 11 genomic districts (PARK1 to 11) related with FPD, 7 were contracted down to single qualities: PARK1 (α-SYNUCLEIN), PARK2 (PARKIN), PARK4 (α- SYNUCLEIN), PARK5 (UCHL1), PARK6 (PINK1), PARK7 (DJ1), PARK8 (DARDARIN/LRRK2), and PARK9 (ATP13A2) [128]. Everything except α-SYNUCLEIN are entirely rationed in the nematode with most deposit positions changed in PD patients encoding indistinguishable amino acids in C. elegans orthologues [129]. Worms overexpressing wild sort, mutant A30P, or A53T human α-SYNUCLEIN in DAergic neurons demonstrate differential levels of damage, including decreased DA content, DAergic neuron degeneration, engine deficiencies reversible by DA organization, intracellular α-SYNUCLEIN totals like Lewy bodies, and expanded helplessness to mitochondrial complex-I inhibitors, which is turned around by treatment with cell reinforcements [130,131]. Moreover, cancellation [132] and knockdown of the C. elegans PARKIN and DJ1 qualities create comparative examples of pharmacological helplessness as those portrayed above for α-SYNUCLEIN overexpression Ved et al. Other PD qualities in C. elegans have been examined. For instance, ubh-1 and ubh-3 [133] have comparative capacities with the human PARK5/UCHL1 orthologue. Ponders on different qualities have been instrumental in disentangling beforehand obscure capacities. For instance, examination of the PARK8/DARDARIN orthologue lrk-1 demonstrated that the protein permits the correct focusing of synaptic vesicle proteins to the axon [134] and ensures against rotenoneinitiated mitochondrial damage Wolozin et al. As of late, RNAi, genomic, and proteomic approaches utilizing human α-SYNUCLEIN transgenic worms distinguished hereditary systems connecting PD to G-protein flagging, endomembrane trafficking, actin cytoskeleton, and oxidative anxiety [135-138], showing the force of this transgenic display for PD think about.

Nonhereditary PD cases have additionally been related with introduction to 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine, an originator medication that is changed over intracerebrally (by astrocytes) to 1-methyl-4-phenylpyridinium (MPP+) by the monoamine oxygenase B. MPP+ harms the DAergic sensory system, prompting to a common Parkinsonian disorder [139,140]. So also, MPP+-uncovered C. elegans indicate particular degeneration of DAergic neurons and related behavioral deformities [141], which is because of ATP consumption Wang et al. Exposures to rotenone (see above) or 6-hydroxydopamine additionally prompt to PD disorders that have comparable components both in people and worms [142-144]. Despite the fact that the nematode does not really show PDlike side effects, comes about with transgenic and medication uncovered worms accentuate the significance of C. elegans as a model creature that (1) grants fast bits of knowledge in the hereditary pathways required in PD and (2) empowers high-throughput screening strategies for the advancement of new against PD drugs [145].

Tauopathies and polyglutamine augmentation issue have likewise been explored in the worm utilizing mutants and transgenic strains [146-150]. The main AD-related proteins distinguished were the betaamyloid peptide forerunner (betaAPP) and the presenilins PS1 and PS2. Investigation of the C. elegans presenilin orthologues sel-12 [151,152] and bounce 1 [153] connected AD to the apoptotic pathway [154] and Notch flagging, which was later affirmed in vertebrates [155,156]. Portrayal of the C. elegans betaAPP orthologue uncovered a key part for microRNA in AD quality control [157]. Be that as it may, the vast majority of the learning about AD gained in C. elegans originated from two transgenic models: worms communicating the human betaAPP [158-162] or TAU [163]. Thinks about on betaAPP transgenic worms uncovered lethality components of AD by distinguishing two new qualities, aph-1 and pen-2, likely required in the movement of the ailment [164]. They additionally permitted the portrayal of oxidation procedures going before fibrillar affidavit and the recognizable proof of qualities endless supply of betaAPP expression [165]. Besides, defensive systems were distinguished [166,167] and potential restorative medications for AD (ginkgolides, Ginkgo biloba separate EGb 761, soy isoflavone glycitein) were initially and effectively examined in worms [168,169]. Caenorhabditis elegans overexpressing the human TAU or a pseudohyperphosphorylated mutant TAU were found to show age-subordinate engine neuron dysfunctions, neurodegeneration, and locomotor deformities because of impeded neurotransmission.

In like manner, while a couple Huntingtin (Htt) - communicating qualities were recognized in C. elegans [170,171], most information originated from transgenic worms communicating polyQ variations of Htt. A few gatherings focused on various neuronal subsets to ponder polyQHtt neurotoxicity in the worm. They depicted behavioral imperfections preceding neurodegeneration and protein collection and axonal deformities and revealed a part for apoptosis in HD neurodegeneration [172,173]. Defensive systems of the polyQ enhancer-1 and ubiquilin were illustrated and pharmacological screening utilizing polyQHtt transgenic C. elegans is continuous.

A last outline of the effective utilization of C. elegans in illustrating the hereditary premise of neurodegenerative issue is exemplified by the portrayal of the hereditary system ensnared in DMD. DMD is mostly described by a dynamic loss of solid mass and capacity happening in guys because of transformations in the DYSTROPHIN quality situated on the X chromosome, which usually prompts to loss of motion and demise by the age of 30. DYSTROPHIN is both solid and neuronal, being required for cerebrum design and neurotransmission, to such an extent that DMD patients show neurodegeneration related with engine shortfalls and diminished subjective exhibitions (normal IQ is 85 in DMD young men) [174-177]. DYSTROPHIN is monitored in C. elegans , however its loss-of-capacity in the worm brings about hypercontractility because of debilitated cholinergic movement and does not influence muscle cells [178,179]. In any case, the perception that twofold mutants for Dystrophin/dys-1 and MyoD/hlh-1 show serious and dynamic muscle degeneration in the worm (as saw in mice), set up the reason for a C. elegans model to examine dystrophinsubordinate myopathies [180]. Utilizing this model, a few accomplices of DYSTROPHIN were described, building up their part in cholinergic neurotransmission and muscle degeneration [181-184]. Also, it was demonstrated that the overexpression of DYSTROBREVIN/dyb-1 postpones neurological and strong imperfections [182], and changes in CHIP/chn-1, substance hindrance of the proteasome, and prednisone or serotonin medicines smother muscle degeneration in C. elegans [185-187].

In this manner, however at first look C. elegans shows up very unique in relation to vertebrates, its anxious hardware and the phone forms managing neuronal advancement, neuronal demise or survival, neurotransmission, and flag combination depend on an indistinguishable neuronal and sub-atomic systems from vertebrates. Joined with the benefits of a little and quickly developing life form, these properties make C. elegans an impeccable framework for quick hereditary examination of neurotoxicity components.

Similar to the case for neurotoxicity, C. elegans gives a practical, in vivo , hereditarily manipulable and physiological model for the investigation of the toxicological results of DNA harm. As portrayed beneath, the hardware that reacts to DNA harm in C. elegans is fundamentally the same as hereditarily to the relating apparatus in higher eukaryotes. Many procedures identified with DNA harm have been widely examined in C. elegans , giving a critical natural setting and clear importance to robotic reviews. At last, intense apparatuses for the investigation of DNA harm, DNA repair, and transformations have been produced in this life form.

Qualities and pathways required in DNA repair in warm blooded animals are by and large all around preserved in C. elegans [188-190]. Proteins required in nucleotide extraction repair, bungle repair, homologous recombination, and nonhomologous end joining, for example, are totally preserved between C. elegans , mouse, and human in view of nucleotide succession homology. This is likewise valid for proteins required in numerous DNA repair–related procedures, for example, translesion DNA polymerases, helicases, and nucleases. Base extraction repair proteins, strangely, demonstrate to some degree less preservation. While this protection is situated now and again just on arrangement homology, large portions of these proteins have now been biochemically or hereditarily described. Fundamentally, proteins required in other DNA harm reactions including apoptosis and cell cycle capture are likewise moderated in C. elegans and well evolved creatures [191].

Early reviews on DNA repair in C. elegans were completed by Hartman and partners, who recognized a progression of radiationtouchy mutants [192,193] and utilized an immune response based examine to quantify acceptance and repair of bright (UV) radiation– induced harm. These and later reviews [194,195] have demonstrated that nucleotide extraction repair is comparative in C. elegans and people both as far as protection of qualities and energy of repair. Nucleotide extraction repair is a basic pathway with regards to introduction to ecological poisons since it perceives and repairs a wide assortment of massive, helix-contorting DNA injuries, including polycyclic sweet-smelling hydrocarbon metabolites, mycotoxins, for example, aflatoxin B1, UV photoproducts, cisplatin adducts, and others [196,197].

While nucleotide extraction repair has been the best-examined DNA repair pathway in C. elegans , critical advance has been made in the investigation of qualities required in other DNA repair pathways also. The part of particular C. elegans quality items in DNA repair has been concentrated both by means of high-throughput and lowthroughput techniques. High-throughput strategies including RNAi knockdown and yeast two-half breed investigation of protein-protein collaboration have been utilized to recognize an expansive number of qualities coding for proteins required in reacting to DNA harm [198,199]. Bring down throughput examines including biochemical examinations of DNA repair exercises [200-206] too in vivo affectability to DNA harming operators [207-212] or other DNA damage–related phenotypes [213-216] have upheld the succession similarity–based recognizable proof of C. elegans homologues of DNA repair qualities in higher vertebrates, and additionally now and again allowing recognizable proof of beforehand obscure qualities required in these pathways.

DNA harm that is not repaired can trigger cell cycle capture and apoptosis, and these pathways are exceptionally very much considered in C. elegans . The immense advance made in comprehension them robotically exhibits the force of this model life form. As said, the cell systems directing apoptosis were found in C. elegans , and apoptosis and cell cycle reactions to DNA harm keep on being intensely considered in C. elegans [217-226]. The short life expectancy of C. elegans has particularly fit momentous reviews on the instruments of germ line eternality [227,228]. Another vital favorable position of C. elegans is the capacity to effortlessly examine in vivo wonders, for example, age-or formative stage–related contrasts in DNA repair limit. For instance, demonstrated that the mistake inclined DNA repair pathway of nonhomologous end joining has practically no part in the repair of DNA twofold strand softens up germ cells however is utilitarian in substantial cells. Demonstrated that checkpoint hushing in light of DNA harm happens in creating incipient organisms however not in the germ line. Both these discoveries are critical in our comprehension formative presentation to genotoxins in that they propose an exceptional security for germ line cells.

DNA damage–related neurotic procedures including carcinogenesis [229-232], maturing [233-239], and neurodegenerative illnesses are additionally ranges of dynamic research in C. elegans . This exploration has both set up the significance of C. elegans as a model for the investigation of genotoxic specialists (because of protection of the DNA harm reaction) and massively expanded its utility in such reviews by giving an abundance of corresponding and relevant natural data identified with the obsessive reactions to DNA harm in this living being.

Caenorhabditis elegans is a fabulous model for investigations of genotoxicity because of the plenty of intense instruments accessible. Hereditary control through RNAi and era of KOs or different mutants is moderately clear. In the event that reasonable mutants are not officially accessible, they can be created by an assortment of methodologies. These incorporate untargeted and focused on techniques, including concoction mutagenesis, transposon inclusion, and biolistic change [240-244].

Tests for the estimation of mutagenesis, DNA harm and repair, and transcriptional movement have additionally been produced for genotoxicity appraisal in C. elegans . Some DNA harm and repair tests in C. elegans can be done with as few as one or a couple of individual nematodes, allowing investigations of interindividual contrasts and allowing high-throughput screening of DNA-harming specialists or qualities required in DNA repair. It is likewise conceivable, utilizing PCR-or Southern blot–based strategies, to recognize harm and repair in various genomic locales and genomes. Mutagenesis has been examined by an assortment of techniques including phenotype-based hereditary transformation inversion screens, an out-of-casing LacZ transgene correspondent, and direct sequencing.

Not at all like the instance of neurotoxicology, there have so far been generally few investigations of genotoxicity in essence utilizing C. elegans (Table 2). One special case has been the investigation of UV radiation, normally as a model genotoxin that presents cumbersome DNA injuries [245-249]. In any case, different classes of genotoxins have been examined, including ionizing radiation [250], substantial metals [251,252], methylmethanesulphonate [253], polycyclic sweetsmelling hydrocarbons (Neher and Sturzenbaum), photosensitizers [254,255], and prooxidant mixes [256,257]. Thinks about have exploited the utility of C. elegans as an in vivo display; for instance, it was demonstrated that nucleotide extraction repair moderated in maturing people and that more extended lived and stress-safe strains have quicker nucleotide extraction repair than do wild sort. It has been conceivable to distinguish cases in which UV resistance was connected to life expectancy, and others in which it was not [258], so that speculations about the relationship of DNA harm and repair with maturing can be straightforwardly tried. Investigations of maturing populaces or people are moderate and costly in mammalian models and unimaginable in vitro .

Endpoint Assay Principle References
A. Mutagenesis Direct sequencing The mutation rate of a given locus is calculated using data from DNA sequencing. Denver et al.(2000, 2004, 2006)
  “Big blue worms” Transgenic C. elegans carrying an out-of-frame LacZ reporter gene expresses blue pigment upon frameshift or insertion/deletion mutations. Pothof et al.(2003); Tijsterman et al. (2002)
  Reversion assay Mutants with an easily scored phenotype (e.g., uncoordinated movement) are exposed to a chemical of interest; the restoration of a normal phenotype indicates mutagenesis. Degtyareva et al. (2002); Greenwald and Horvitz (1980); Hartman et al. (1995)
  Lethality assay The lethality of transgenic, mutation-sensitive C. elegans was measured for mutagen detection Rosenbluth et al. (1983); Rosenbluth et al. (1985)
B. DNA damage and repair PCR-based assay The amount of PCR product is inversely proportional to the amount of DNA damage on a given length of template Meyer et al.(2007); Neher and Sturzenbaum (2006)
  Southern blot T4 endonuclease–sensitive sites in specific genes (identified by genomic DNA sequence) indicate the presence of UV photodimers Hyun et al.(2008)
  Immunoassay Antibodies to specific UV photoproducts are identified Hartman et al. (1989)
  Enzymatic activity A diagnostic enzymatic activity is measured in vitro Shatilla and Ramotar (2002)
  Reproduction/development assay with KO mutants Specific DNA damage (e.g., DNA adduct) can be tested using simple reproduction/development assays with mutants lacking a specific DNA repair pathway (e.g., nucleotide excision repair) Parket al.(2002, 2004)
C. Transcriptional activities RNA: DNA ratio A decrease in RNA: DNA ratio indicates the inhibition of transcriptional activities Ibiam and Grant (2005)

Table 2: Genotoxicity assays available for the caenorhabditis elegans model.

High-throughput screening has two particular definitions in toxicology: (1) far reaching screens for atomic targets or go betweens of harmfulness and (2) quick, high-content substance screens to recognize potential toxicants. An all-inclusive screen can fill in as a speculation discovering instrument, giving a bearing to encourage robotic examination. This approach is especially valuable for concentrate any toxicant with an inadequately comprehended system of activity. Expansive screens should be possible utilizing forward hereditary qualities, DNA microarrays, or all-inclusive RNAi in C. elegans .

High-throughput synthetic screening, in examination, has been proposed as a speedier and less costly technique for harmfulness testing [259]. The routine creature testing utilized by organizations or offices is work concentrated and tedious, bringing about an extensive number of toxicants not being tried by any means. It is assessed, for example, that there are more than 10,000 ecological chemicals from a few Environmental Protection Agency programs that require additionally testing [260]. The goal of high-throughput compound screening is to waitlist chemicals demonstrating high danger, subsequently setting need for controls and also advance lethality testing in mammalian models.

High-throughput screening is plausible with C. elegans because of its test manipulability and also a few robotization advances. Caenorhabditis elegans is anything but difficult to deal with in the research center; it can be developed on strong support or in fluid, in Petri dishes, tubes, or 6-, 12-, 24-, 96-, or 384-well plates. It can likewise be presented to toxicants intensely or constantly by infusion, bolstering, or drenching. Robotized imaging techniques for absorbance, fluorescence, development, or morphometric estimation have been created since the late 1980s [261-266]. These days, cell sorters adjusted to sort worms in view of morphometric parameters or articulation of fluorescent proteins consolidated with imaging stages have been effectively utilized for expansive scale promoter expression examinations and medication screening purposes [267,268]. As of late, a microfluidic C. elegans sorter with three dimensional subcellular imaging capacities was created, permitting high-throughput measures of higher intricacy [269].

While the straightforwardness and manipulability of the C. elegans framework empowers high-throughput approaches, it additionally prompts to a few potential burdens in toxicology thinks about. Caenorhabditis elegans displays vital metabolic contrasts contrasted with vertebrates. For instance, C. elegans is exceptionally impervious to benzo[a]pyrene [270], likely on the grounds that it doesn't metabolize the concoction. This issue can be possibly comprehended, in any case, by communicating the vertebrate cytochrome P450s in C. elegans . The impermeable fingernail skin layer and additionally particular intestinal take-up, besides, may hinder the section of chemicals, in this way requiring high introduction dosages to affect the worm's physiology. A mutant strain (dal-1) has as of late been detached that is sound under lab conditions however displays adjusted intestinal morphology and expanded intestinal ingestion of an extensive variety of medications. The resultant-expanded helplessness of this strain to the poisonous or pharmacological exercises of tried mixes can possibly build the affectability of the C. elegans framework.

Forward hereditary qualities allude to the investigation of qualities in view of a given phenotype. In a forward hereditary qualities screen, C. elegans are treated with a mutagen, as portrayed previously. Mutant strains are then presented to a toxicant and are screened for expanded resistance or affectability. Once a safe or extremely touchy mutant is distinguished, the change is found utilizing two-point and three-point mapping and affirmed utilizing single-quality save or RNAi phenocopying. Forward hereditary qualities are productive in C. elegans in light of the fact that the mutants can cover qualities communicated in an assortment of tissues. Caenorhabditis elegans is bisexual, so homozygous mutant strains can be created in the F2 era by means of self-intersection.

Forward hereditary qualities screens are a helpful strategy in unthinking toxicology [271,272]. For example, found the part of glycolipid receptors and sugar digestion system in Bacillus thuringiensis (Bt) poisons utilizing C. elegans subjected to a forward hereditary qualities screen. The transformation of glycolipid receptors keeps Bt poison from entering intestinal epithelium in C. elegans . Such a tissue-particular component would have been hard to identify utilizing as a part of vitro cell societies.

Caenorhabditis elegans has a few points of interest over different species in quality expression investigation [273], the data rich focal genomic database of C. elegans , gives an instinctive interface into a very much explained genome. Caenorhabditis elegans likewise has a steady arrangement of quality distinguishing proof, along these lines keeping away from the disarray of quality recognizable proof that is regular in numerous species, including human. The interactome demonstrating of C. elegans is likewise the most created among every creature specie and alongside other genome-level bioinformatics instruments [274] enormously encourages framework based investigation.

The aftereffects of quality expression examination can be approved in vivo utilizing mutational or transgenic approaches in C. elegans (Tables 3 and 4). For instance, the quality articulation of C. elegans presented to ethanol, atrazine, polychlorinated biphenyls, endocrine upsetting chemicals, and polycyclic sweet-smelling hydrocarbons have been profiled [275,276]. Line up studies with transgenic C. elegans communicating fluorescent markers were utilized to identify overexpression of protein in particular tissues in vivo . Mutant C. elegans were likewise used to affirm the part of particular sub-atomic targets in view of quality expression investigation.

Approach/toxin investigated Mutants used Major findings References
A. KO mutant analysis
Black widow spider venom lat-1: KO of latrophilin Latrophilin is the receptor responsible for the toxicity of venom Mee et al.(2004)
As asna-1: KO of ArsA ATPase ArsA ATPase is important in Ar resistance in both bacteria and animals Tseng et al.(2007)
Cd pgp-5: KO of a ABC transporter ABC transporter is required for resistance to Cd toxicity Kurz et al.(2007)
PCB52 cyp-35A1 to cyp-35A5: KOs of cytochrome P450 35A subfamily CYP35A is required for fat storage and resistance to PCB52 toxicity Menzel et al.(2007)
B. Forward genetics screen
BPA bis-1: mutant created from EMS mutagenesis Collagen mutants are hypersensitive to BPA Watanabe et al.(2005)
Phosphine pre-1, pre-7, pre-33: mutants created from EMS mutagenesis Uptake and oxidization of phosphone are directly associated with oxidative stress in cells Cheng et al.(2003)
Bt toxins bre-1 to bre-5: mutants created from EMS mutagenesis Five new genes involved in Bt toxicity are identified Marroquin et al. (2000)
bre-5: KO of β-1,3-galactosyltransferase Carbohydrate modification is involved in Bt toxicity Griffitts et al. (2001)
bre-2 to bre-5: KOs of glycolipid carbohydrate metabolism Glycolipid receptors are targets of Bt toxins Griffitts et al. (2005)
bre-1: KO of GDP-mannose 4,6-dehydratase The monosaccharide biosynthetic pathway is involved in Bt toxicity Barrows et al. (2007b)

Table 3: Examples of Mutational Analysis of Caenorhabditis elegans in Toxicology Research. Note: ABC, ATP-binding cassette; PCB52, polychlorinated biphenyl 52; EMS, ethane methyl sulfonate.

Field/target tagged Reporter used Applications References
A. Mechanistic studies
DAergic neurons GFP Detect neurodegradation caused by chemicals Jiang et al.(2007)
CYP14A3 and 35A3 GFP Detect intestinal CYP overexpression in response to PCB52 as well as other xenobiotic CYP inducers Menzel et al.(2007)
GST GFP Measure GST induction in response to acrylamide as well as other inducers of oxidative stress Hasegawa and van der Bliek (in press)
B. Environmental biomonitoring
Heat shock proteins GFP; β-galactosidase Widely used for measuring stress response associated to toxicity of heavy metals, fungicides, pharmaceuticals, as well as field samples Dengg and van Meel (2004); Easton et al. (2001); Mutwakil et al.  (1997); Roh et al (2006)
Metallothionein β-galactosidase Specifically used for monitoring the bioavailability of heavy metals Cioci et al.(2000)
ATP level Firefly luciferase Measure the reduction of metabolic activity in response to environmental stressor Lagido et al.2001

Table 4: Examples of Transgenic Caenorhabditis elegans Used in Toxicology Research. Note: CYP, cytochrome P450; GST, glutathione Stransferase.

The disclosure of RNAi systems in C. elegans for which the 2006 Nobel Prize was granted [277,278] and the total sequencing of the nematode genome (C. elegans Sequencing Consortium) prompted to the era of publically accessible RNAi libraries covering 90% of its qualities [279-281]. Methodologies to enhance RNAi proficiency, particularly in neurons, were further created [282-285]. RNAi can be activated by infusion of worms with meddling twofold strand RNA (dsRNA), by nourishing them with transgenic microscopic organisms creating the dsRNA or by absorbing them an answer of dsRNA. The last permit coordinated RNAi introduction and broad screens in 96-or 384-well plates with fluid worm societies and have added to disclosures of components of axon direction and also mitochondrial contribution in oxidative anxiety and maturing [286-290].

A broad RNAi screen commonly surveys various physiological parameters in the meantime, for example, suitability, development, nourishment admission, and advancement, along these lines encouraging the elucidation of screening results. While most RNAi screens have been done in wild-sort C. elegans , some are performed utilizing KO mutants to give touchier or particular measures (Kaletta and Hengartner). Vast RNAi screens are turning into a technique for decision for finding quality capacity. A current review by Kim and Sun, for instance, distinguished various daf-2-needy and supplement responsive qualities that are receptive to paraquat-actuated oxidative anxiety.

The utilization of C. elegans as a prescient model for human danger was initially proposed with regards to substantial metals [291]. The C. elegans measure was approved as an indicator of mammalian intense lethality utilizing eight diverse metal salts, producing LC50 values parallel to the rodent and mouse LD50 values. A later review researched the intense behavioral harmfulness of 15 OP pesticides in C. elegans [292]. The harmfulness of these pesticides in C. elegans was observed to be essentially connected to the LD50 intense lethality values in rats and mice (Table 5). A few different reviews have additionally approved various C. elegans –based examines for foreseeing neurological and formative danger in mammalian species [293,294].

Compound Strains investigated Observations References
Paraquat mev-1(kn1)a, mev-2(kn2)a Hypersensitive to oxygen and paraquat, decreased SOD activityb Ishii et al.(1990)
rad-8(mn162) Hypersensitive to oxygen and paraquat, reduced fecundity, decreased life span Ishii et al.(1993)
age-1(hx542), age-1(hx546) Increased catalase and Cu/Zn SOD activity, increased life span Vanfleteren (1993)
Vitamin E (antioxidant) inhibits oxidative damage from paraquatb Goldstein and Modric (1994)
mev-1(kn1), rad-8(mn162) Paraquat and high oxygen content inhibit development, inversely proportional to life span Hartman et al. (1995)
age-1(hx546), daf-16(m26), mev-1(kn1)a Increased resistance to paraquat and heat, extended life span, increased SOD, and catalase mRNA level only in age-1 mutant, but not daf-16 or mev-1 Yanase et al.(2002)
mev-5(qa5005)a, mev-6(qa5006)a, mev-7(qa5007)a Longevity and sensitivity to paraquat, UV or heat do not correlate Fujii et al.(2005)
mev-1(kn1), gas-1(fc21) Overproduction of superoxide anion in submitochondrial particles upon paraquat exposure Kondo et al.(2005)
skn-1(zu67) Activation of SKN-1 transcription factor, localizes to the nucleus following paraquat exposure Kell et al.(2007)
daf-2(e1370) Extended animal life span and increased resistance to ROS produced by paraquat Kim and Sun (2007); Yang et al.(2007)
Overexpression of GSTO, gsto-1RNAi Increased resistance to paraquat-induced oxidative stress Burmeister et al. (2008)
Rotenone  gas-1(fc21) Increased sensitivity to rotenone under hyperoxia Ishiguro et al.(2001)
pdr-1, djr-1.1RNAi Increased vulnerability to rotenone Ved et al.(2005)
Overexpression of LRRK2, lrk-1RNAi Overexpression of wild-type LRRK2 strongly protects against rotenone toxicity Wolozin et al. (2008)
Ops N2 Computer tracking system is a promising tool for assessing neurobehavioral changes associated with OP toxicity Williams and Dusenbery 1990
Cholinesterase inhibition associated with high behavioral toxicity Cole et al.(2004)
Absorption effects are more prominent than biodegradation in soil toxicity tests Saffih-Hdadi et al.(2005)
Carbamates N2 Rank order of toxicity of carbamate pesticides in C. elegans correlates well with values for rats and mice, and degree of behavioral alteration correlates with AChE inhibition Melstrom and Williams (2007)
Bt toxin bre-1(ye4), bre-2(ye31), bre-3(ye28), bre-4(ye13), bre-5(ye17) Extensive damage to gut, decreased fertility, and death Marroquin et al. (2000)
bre-5(ye17) Increased resistance tonBtntoxin Griffittsnet al. (2001)
bre-2(ye31), bre-2(ye71),bre-3(ye28), bre-4(ye13) Bt toxin resistance involves the loss of glycosyltransferase in the intestine Griffitts et al. (2003)
glp-4(bn2), kgb-1(um3), jnk-1(gk7), sek-1(km4) Bt toxin reduces brood size and causes damage to the intestine Wei et al.(2003)
A p38 MAPK and a c-Jun N-terminal-like MAPK are both transcriptionally upregulated by Bttoxin Huffman et al. (2004a, 2004b)
Survival rate, infection level, and behavior differred in C. elegans isolated from geographically distinct strains Schulenburg and Muller (2004)
bre-2(ye31), bre-3(ye28), bre-4(ye13), bre-5(ye17) Bt toxin resistance entails loss of glycolipid carbohydrates and the toxin directly and specifically binds to Glycolipids Griffitts et al. (2005)
bre-3(ye28) Resistance to Bt toxin develops as a result of loss of glycolipid receptors for the toxin Barrows et al. (2006)
bre-1(ye4), bre-2(ye31) Resistance to toxin is achieved by mutations in gylcosyltransferase genes that glycosylate glycolipid or with a loss of the monosaccharide biosynthetic pathway Barrows et al. (2007a, 2007b)
daf-2(e1370), daf-2(e1368), age-1(hx546), daf-16(mgDf50), daf-2(0(m26) Mutations in the insulin-like receptor pathway lead to distinct behavioral responses, including the evasion of pathogens and reduced ingestion Hasshoff et al. (2007) 
Reproduction and growth significantly reduced by Bt toxin Hoss et al.(2008)
Captan hsp-16.48;hsp-16.1:lacZ Stress induction localized to muscle cells of the pharynx Jones et al.(1996)
Inhibits feeding, cessation of muscular contraction
Dithiocarbamate fungicides hsp-16.48;hsp-16.1:lacZ Induction of stress response Guven et al.(1999)
Organochlorinated pesticides N2 Decreased sensitivity to organochlorinated pesticide in C. elegansthan other soil invertebrates. Compared to other organic pollutants tested, organochlorinated pesticides are the most toxic substances in soil or aquatic medium Bezchlebova et al.(2007); Sochova et al. (2007)

Table 5: Pesticides that Have Been Tested Using Caenorhabditis elegans as a Model Organism. Note. MAPK, mitogen-activated protein kinase; ROS, reactive oxygen species. aThese mutants showed defective dye filling, indicative of chemosensory neuron damage.bSOD, superoxide dismutase.

A C. elegans –based, high-throughput poisonous quality screen was initially distributed by the Freedman aggregate at National Institute of Environmental Health Sciences [295]; extra gatherings including industry and government assembles in the United States and somewhere else are additionally doing high-throughput danger screening. Screens are ordinarily directed on a 96-well plate with a mechanical fluid taking care of workstation (Biosort, Union Biometrica, Inc., Holliston, MA) to investigate the length, optical thickness, movement, and fluorescence of C. elegans . Caenorhabditis elegans is refined in fluid from treated egg to grown-up through four unmistakable larval stages. The advancement, generation, and encouraging practices of the C. elegans culture because of various synthetic exposures are described. The screen has been approved by the Freedman bunch with 60 chemicals including metals, pesticides, mutagens, and nontoxic specialists [295].

The high-throughput poisonous quality screen is as a rule additionally enhanced with extra hereditary qualities and mechanization systems. The summed up stress reaction of C. elegans , for example, was pictured with transgenic GFP builds, giving a more delicate end point for lethality screens [296]. Nematode velocity can be followed consequently, giving a more touchy screen of neurotoxicity. Transgenic or mutant C. elegans can likewise be utilized as a part of the high-throughput screen to identify particular methods of activity, including metal reaction [297], oxidative anxiety [298,299], and DNA harm [300]. A microfluidic C. elegans sorter with three-dimensional subcellular imaging capacities was as of late revealed, subsequently permitting high-throughput examines of higher many-sided quality.

Nematodes are the most plenteous creature in soil biological communities and furthermore found in sea-going and silt situations. They serve numerous imperative parts in supplement cycling and in keeping up natural quality. These components have upheld their utilization in ecotoxicological considers and, from the late 1970s, an assortment of nematode animal categories have been utilized to concentrate ecological issues. Amid the late 1990s, C. elegans started to rise as the nematode types of decision in light of the enormous assortment of learning created by essential researchers utilizing this model living being for organic reviews. Albeit for the most part considered a dirt life form, C. elegans lives in the interstitial water between soil particles and can be effectively refined inside the research center in amphibian medium. The larger part of natural reviews has been performed in an oceanic medium, given its usability, and as toxicological end focuses have been produced, the evaluation apparatuses have been connected to residue and soil medium which takes into consideration a more important direct ecological correlation.

The ecological toxicological writing utilizing C. elegans is broad and gives an outline of research center based reviews where a toxicant of natural intrigue has been added to a medium (water, dregs, or soil) trailed by presentation to C. elegans and the evaluation of an unfriendly impact. In a set number of circumstances, C. elegans testing has been utilized to evaluate defilement in field settings. A significant part of the early work investigated metal poisonous quality and utilized lethality as an endpoint. After some time, a more extensive assortment of toxicants has been tried and more refined sublethal end focuses have been produced including the utilization of transgenic strains with particular biomarkers [301-305], development and propagation [306], nourishing [307] and development. These sorts of end focuses created through natural reviews are specifically relevant to the utilization of the life form as an option for mammalian testing.

Two of the main restrictions in utilizing C. elegans in ecological testing are concerns identified with its correlation with different nematodes and solid and basic techniques for separating them from soil and dregs. Given the practically incalculable assortment of nematodes, it is unimaginable for one animal group to be illustrative of the whole Nematoda phylum. Constrained reviews looking at the toxicological impacts between nematodes species show that C. elegans is as delegate as any of the ones generally utilized and, as a rule, little contrast accordingly has been found between species [308-314]. Advance, this creature is considerably more altogether comprehended and advantages from its convenience.

Much advance has been improved to create techniques to remove the worm from soil and silt. The underlying strategy created by Donkin and Dusenbery [315-328] has prompted to an institutionalized soil toxicological testing technique received in 2001 by the American Society for Testing and Materials [329-353] and as of late the International Standards Organization in Europe (ISO 2007). The underlying extraction technique has been enhanced using transgenic strains of nematodes which takes into consideration GFP-named worms to be utilized that recognizes the worms being tried in soils from the vast quantities of indigenous species that are comparable in size and appearance. It additionally makes less demanding expulsion from soil with high natural substance. This work has prompted to more enthusiasm for utilizing C. elegans in ecological reviews.

The extraordinary elements of C. elegans [353-373] make it a brilliant model to supplement mammalian models in toxicology look into. Tries different things with C. elegans don't cause an indistinguishable expense from investigations with in vivo vertebrate models, while as yet allowing testing of speculations in an in place metazoan life form. The hereditary apparatuses accessible for C. elegans make it a superb model for concentrate the parts of particular qualities in toxicological procedures and quality environment associations, while the life history of this life form fits high-throughput examinations. Therefore, C. elegans speaks to a brilliant supplement to in vitro or cell culture–based frameworks and in vivo vertebrate models.

References

  1. Hodgkin J, Hope IA (1999) C. elegans: A Practical Approach. WormBook 245-270.
  2. Caswell-Chen EP, Chen J, Lewis EE, Douhan GW, Nadler SA, et al. (2005) Revising the standard wisdom of C. elegans natural history: Ecology of longevity. Sci Aging Knowledge Environ 2005: 30.
  3. Kiontke K, Sudhaus W (2006) Ecology of Caenorhabditis species. WormBook.
  4. Maupus E (1900) Modes et formes de reproduction des nematodes. Arch Zool Exp Gen 8: 463-624.
  5. Antoshechkin I, Sternberg PW (2007) The versatile worm: genetic and genomic resources for Caenorhabditis elegans research. Nat Rev Genet 8: 518-532.
  6. Kaletta T, Hengartner MO (2006) Finding function in novel targets: C. elegans as a model organism. Nat Rev Drug Discov 5: 387-398.
  7. Brenner S (1974) The genetics of Caenorhabditis elegans. Genetics 77: 71-94.
  8. Sulston JE (1983) Neuronal cell lineages in the nematode Caenorhabditis elegans. Cold Spring Harb Symp Quant Biol 48 Pt 2: 443-452.
  9. Sulston JE, Schierenberg E, White JG, Thomson JN (1983) The embryonic cell lineage of the nematode Caenorhabditis elegans. Dev Biol 100: 64-119.
  10. Chen BL, Hall DH, Chklovskii DB (2006) Wiring optimization can relate neuronal structure and function. Proc Natl Acad Sci U S A 103: 4723-4728.
  11. Brockie PJ, Maricq AV (2006) Ionotropic glutamate receptors: genetics, behavior and electrophysiology. WormBook.
  12. Avery L, Horvitz HR (1990) Effects of starvation and neuroactive drugs on feeding in Caenorhabditis elegans. J Exp Zool 253: 263-270.
  13. Chase DL, Koelle MR (2007) Biogenic amine neurotransmitters in C. elegans. WormBook.
  14. Morgan PG, Kayser EB, Sedensky MM (2007) C. elegans and volatile anesthetics. WormBook .
  15. Bianchi L, Driscoll M (2006) Heterologous expression of C. elegans ion channels in Xenopus oocytes. WormBook.
  16. Cook A, Franks CJ, Holden-Dye L (2006) Electrophysiological recordings from the pharynx. WormBook.
  17. Schafer WR (2006) Neurophysiological methods in C. elegans: an introduction. WormBook.
  18. Chisholm AD, Jin Y (2005) Neuronal differentiation in C. elegans. Curr Opin Cell Biol 17: 682-689.
  19. Silhankova M, Korswagen HC (2007) Migration of neuronal cells along the anterior-posterior body axis of C. elegans: Wnts are in control. Curr Opin Genet Dev 17: 320-325.
  20. Quinn CC, Wadsworth WG (2006) Axon guidance: ephrins at WRK on the midline. Curr Biol 16: R954-955.
  21. Wadsworth WG (2002) Moving around in a worm: netrin UNC-6 and circumferential axon guidance in C. elegans. Trends Neurosci 25: 423-429.
  22. Jin Y (2002) Synaptogenesis: insights from worm and fly. Curr Opin Neurobiol 12: 71-79.
  23. Gabel CV, Antonie F, Chuang CF, Samuel AD, Chang C (2008) Distinct cellular and molecular mechanisms mediate initial axon development and adult-stage axon regeneration in C. elegans. Development 135: 1129-1136.
  24. Wu Z, Ghosh-Roy A, Yanik MF, Zhang JZ, Jin Y, et al. (2007) Caenorhabditis elegans neuronal regeneration is influenced by life stage, ephrin signaling, and synaptic branching. Proc Natl Acad Sci U S A 104: 15132-15137.
  25. Horvitz HR (2003) Worms, life, and death (Nobel lecture). Chembiochem 4: 697-711.
  26. Sulston JE (2003) Caenorhabditis elegans: the cell lineage and beyond (Nobel lecture). Chembiochem 4: 688-696.
  27. Bharathi, Ravid R, Rao KS (2006) Role of metals in neuronal apoptosis: challenges associated with neurodegeneration. Curr Alzheimer Res 3: 311-326.
  28. Hirata Y (2002) Manganese-induced apoptosis in PC12 cells. Neurotoxicol Teratol 24: 639-653.
  29. Mattson MP (2000) Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol 1: 120-129.
  30. Ong WY, Farooqui AA (2005) Iron, neuroinflammation, and Alzheimer's disease. J Alzheimers Dis 8: 183-200.
  31. Savory J, Herman MM, Ghribi O (2003) Intracellular mechanisms underlying aluminum-induced apoptosis in rabbit brain. J Inorg Biochem 97: 151-154.
  32. Ayyadevara S, Engle MR, Singh SP, Dandapat A, Lichti CF, et al. (2005) Lifespan and stress resistance of Caenorhabditis elegans are increased by expression of glutathione transferases capable of metabolizing the lipid peroxidation product 4-hydroxynonenal. Aging Cell 4: 257-271.
  33. Ayyadevara S, Alla R, Thaden JJ, Shmookler Reis RJ (2008) Remarkable longevity and stress resistance of nematode PI3K-null mutants. Aging Cell 7: 13-22.
  34. Daitoku H, Fukamizu A (2007) FOXO transcription factors in the regulatory networks of longevity. J Biochem 141: 769-774.
  35. Gami MS, Iser WB, Hanselman KB, Wolkow CA (2006) Activated AKT/PKB signaling in C. elegans uncouples temporally distinct outputs of DAF-2/insulin-like signaling. BMC Dev Biol 6: 45.
  36. Grad LI, Lemire BD (2004) Mitochondrial complex I mutations in Caenorhabditis elegans produce cytochrome C oxidase deficiency, oxidative stress and vitamin-responsive lactic acidosis. Hum Mol Genet 13: 303-314.
  37. Inoue H, Hisamoto N, An JH, Oliveira RP, Nishida E, et al. (2005) The C. elegans p38 MAPK pathway regulates nuclear localization of the transcription factor SKN-1 in oxidative stress response. Genes Dev 19: 2278-2283.
  38. Leiers B, Kampkotter A, Grevelding CG, Link CD, Johnson TE, et al. (2003) A stress-responsive glutathione S-transferase confers resistance to oxidative stress in Caenorhabditis elegans. Free Radic. Biol Med 34: 1405-1415
  39. Tullet JM, Hertweck M, An JH, Baker J, Hwang JY, et al. (2008) Direct inhibition of the longevity-promoting factor SKN-1 by insulin-like signaling in C. elegans. Cell 132: 1025-1038.
  40. Wang XW, Liou YC, Ho B, Ding JL (2007) An evolutionarily conserved 16-kDa thioredoxin-related protein is an antioxidant which regulates the NF-kappaB signaling pathway. Free Radic Biol Med 42: 247-259.
  41. Inoue T, Thomas JH (2000) Suppressors of transforming growth factor-beta pathway mutants in the Caenorhabditis elegans dauer formation pathway. Genetics 156: 1035-1046.
  42. Malone EA, Thomas JH (1994) A screen for nonconditional dauer-constitutive mutations in Caenorhabditis elegans. Genetics 136: 879-886.
  43. Mörck C, Axäng C, Pilon M (2003) A genetic analysis of axon guidance in the C elegans pharynx. Dev Biol 260: 158-175.
  44. Nass R, Hahn MK, Jessen T, McDonald PW, Carvelli L, et al. (2005) A genetic screen in Caenorhabditis elegans for dopamine neuron insensitivity to 6-hydroxydopamine identifies dopamine transporter mutants impacting transporter biosynthesis and trafficking. J Neurochem 94: 774-785.
  45. O'Connell KF, Leys CM, White JG (1998) A genetic screen for temperature-sensitive cell-division mutants of Caenorhabditis elegans. Genetics 149: 1303-1321.
  46. Houthoofd K, Braeckman BP, Lenaerts I, Brys K, De Vreese A, et al. (2002) Ageing is reversed, and metabolism is reset to young levels in recovering dauer larvae of C. elegans. Exp Gerontol 37: 1015-1021.
  47. Anderson GL, Boyd WA, Williams PL (2001) Assessment of sublethal endpoints for toxicity testing with the nematode Caenorhabditis elegans. Environ Toxicol Chem 20: 833-838.
  48. Johnson TE, Nelson GA (1991) Caenorhabditis elegans: a model system for space biology studies. Exp Gerontol 26: 299-309.
  49. Boyd WA, Cole RD, Anderson GL, Williams PL (2003) The effects of metals and food availability on the behavior of Caenorhabditis elegans. Environ Toxicol Chem 22: 3049-3055.
  50. Williams PL, Dusenbery DB (1988) Using the nematode Caenorhabditis elegans to predict mammalian acute lethality to metallic salts. Toxicol Ind Health 4: 469-478.
  51. Wang S, Tang M, Pei B, Xiao X, Wang J, et al. (2008) Cadmium-induced germline apoptosis in Caenorhabditis elegans: the roles of HUS1, p53, and MAPK signaling pathways. Toxicol Sci 102: 345-351.
  52. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Richardson RJ (1998) The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living. Neurology 50: 1346-1350.
  53. Boyd WA, Williams PL (2003) Comparison of the sensitivity of three nematode species to copper and their utility in aquatic and soil toxicity tests. Environ Toxicol Chem 22: 2768-2774.
  54. Candido EP, Jones D (1996) Transgenic Caenorhabditis elegans strains as biosensors. Trends Biotechnol 14: 125-129.
  55. Riddle DL, Blumenthal T, Meyer BJ, Preiss JR (1997) C. elegans II. , 1997Cold Spring Harbor, NYCold Spring Harbor Laboratory Press.
  56. Williams PL, Anderson GL, Johnstone JL, Nunn AD, Tweedle MF, et al. (2000) Caenorhabditis elegans as an alternative animal species. J Toxicol Environ Health A 61: 641-647.
  57. Hartman PS, Simpson VJ, Johnson T, Mitchell D (1998) Radiation sensitivity and DNA repair in Caenorhabditis elegans strains with different mean life spans. Mutat Res 208: 77-82.
  58. Hilliard MA, Apicella AJ, Kerr R, Suzuki H, Bazzicalupo P, et al. (2005) In vivo imaging of C. elegans ASH neurons: cellular response and adaptation to chemical repellents. EMBO J 24: 63-72.
  59. Rankin CH, Beck CD, Chiba CM (1990) Caenorhabditis elegans: a new model system for the study of learning and memory. Behav Brain Res 37: 89-92.
  60. Mori I, Sasakura H, Kuhara A (2007) Worm thermotaxis: a model system for analyzing thermosensation and neural plasticity. Curr Opin Neurobiol 17: 712-719.
  61. Ye H, Ye B, Wang D (2008) Trace administration of vitamin E can retrieve and prevent UV-irradiation and metal exposure-induced memory deficits in nematode Caenorhabditis elegans. Neurobiol Learn Mem 90: 10-18.
  62. Garza A, Vega R, Soto E (2006) Cellular mechanisms of lead neurotoxicity. Med Sci Monit 12: RA57-65.
  63. Goncalves PP, Silva VS (2007) Does neurotransmission impairment accompany aluminium neurotoxicity? J Inorg Biochem 101: 1291-1338.
  64. Liao VH, Yu CW (2005) Caenorhabditis elegans gcs-1 confers resistance to arsenic-induced oxidative stress. Biometals 18: 519-528.
  65. Atchison WD (2003) Effects of toxic environmental contaminants on voltage-gated calcium channel function: From past to present. J Bioenerg Biomembr 35: 507-532.
  66. Koselke L, Sam C, Hajela R, Atchison B (2007) Protective effects of verapamil on mercury toxicity in C. elegans. Toxicol Sci 106: 5-28.
  67. Jiang GC, Tidwell K, McLaughlin BA, Cai J, Gupta RC, et al. (2007) Neurotoxic potential of depleted uranium effects in primary cortical neuron cultures and in Caenorhabditis elegans. Toxicol Sci 99: 553-565.
  68. Kim SK, Lund J, Kiraly M, Duke K, Jiang M, et al. (2001) A gene expression map for Caenorhabditis elegans. Science 293: 2087-2092.
  69. Cui Y, Boyd WA, McBride SJ, Freedman JH (2007) Toxicogenomic analysis of Caenorhabditis elegans reveals novel genes and pathways involved in the resistance to cadmium toxicity. Genome Biol 8: R122.
  70. Cui YX, McBride SJ, Boyd WA, Alper S, Freedman JH (2007) Toxicogenomic analysis of Caenorhabditis elegans reveals novel genes and pathways involved in the resistance to cadmium toxicity. Genome Biol 8: R122
  71. Jackson D, Lewis J, Anderson S, Gehman E, Szilagyi M, et al. (2006) A nematode model to elucidate mechanisms of developmental toxicity. Toxicol Sci 81: 390-400.
  72. Tvermoes B, Freedman JH (2008) Caenorhabditis elegans gene, Numr-1, assembles into nuclear stress granules after cadmium treatment, SOT Meeting March 16-20. Society of Toxicology 331–332 .
  73. Semchuk KM, Love EJ, Lee RG (1992) Parkinson's disease and exposure to agricultural work and pesticide chemicals. Neurology 42: 1328-1335.
  74. Ritz B, Yu F (2000) Parkinson's disease mortality and pesticide exposure in California 1984-1994. Int J Epidemiol 29: 323-329.
  75. Thiruchelvam M, Brockel BJ, Richfield EK, Baggs RB, Cory-Slechta DA (2000) Potentiated and preferential effects of combined paraquat and maneb on nigrostriatal dopamine systems: Environmental risk factors for Parkinson's disease? Brain Res 873: 225-234.
  76. Hills T, Brockie PJ, Maricq AV (2004) Dopamine and glutamate control area-restricted search behavior in Caenorhabditis elegans. J Neurosci 24: 1217-1225.
  77. Sanyal S, Wintle RF, Kindt KS, Nuttley WM, Arvan R, et al. (2004) Dopamine modulates the plasticity of mechanosensory responses in Caenorhabditis elegans. EMBO J 23: 473-482.
  78. Sawin ER, Ranganathan R, Horvitz HR (2000) C. elegans locomotory rate is modulated by the environment through a dopaminergic pathway and by experience through a serotonergic pathway. Neuron 26: 619-631.
  79. Hartman P, Childress E, Beyer T (1995) Nematode development is inhibited by methyl viologen and high oxygen concentrations at a rate inversely proportional to life span. J Gerontol A Biol Sci Med Sci 50: B322-B6.
  80. Kondo M, Senoo-Matsuda N, Yanase S (2005) Effect of oxidative stress on translocation of DAF-16 in oxygen-sensitive mutants, mev-1 and gas-1 of Caenorhabditis elegans. Mech Ageing Dev 126: 637-641
  81. Yamamoto K, Honda S, Ishii N (1996) Properties of an oxygen-sensitive mutant mev-3 of the nematode Caenorhabditis elegans. Mutat Res 358: 1-6.
  82. Guo J, Lemire BD (2003) The ubiquinone-binding site of the Saccharomyces cerevisiae succinate-ubiquinone oxidoreductase is a source of superoxide. J Biol Chem 278: 47629-47635
  83. Ishii N, Takahashi K, Tomita S, Keino T, Honda S, et al. (1990) A methyl viologen-sensitive mutant of the nematode Caenorhabditis elegans. Mutat Res 237: 165-171.
  84. Fujii M, Matsumoto Y, Tanaka N, Miki K, Suzuki T, et al. (2004) Mutations in chemosensory cilia cause resistance to paraquat in nematode Caenorhabditis elegans. J Biol Chem 279: 20277-20282.
  85. Fujii M, Tanaka N, Miki K, Hossain MN, Endoh M, et al. (2005) Uncoupling of longevity and paraquat resistance in mutants of the nematode Caenorhabditis elegans. Biosci Biotechnol Biochem 69: 2015-2018.
  86. Yang W, Li J, Hekimi S (2007) A measurable increase in oxidative damage due to reduction in superoxide detoxification fails to shorten the life span of long-lived mitochondrial mutants of Caenorhabditis elegans. Genetics 177: 2063-2074.
  87. Vanfleteren JR (1993) Oxidative stress and ageing in Caenorhabditis elegans. Biochem J 292: 605-608.
  88. Yanase S, Yasuda K, Ishii N (2002) Adaptive responses to oxidative damage in three mutants of Caenorhabditis elegans (age-1, mev-1 and daf-16) that affect life span. Mech Ageing Dev 123: 1579-1587.
  89. Burmeister C, Luersen K, Heinick A, Hussein A, Domagalski M, et al. (2008) Oxidative stress in Caenorhabditis elegans: Protective effects of the Omega class glutathione transferase (GSTO-1). FASEB J 22: 343-354.
  90. Honda S, Ishii N, Suzuki K, Matsuo M (1993) Oxygen-dependent perturbation of life span and aging rate in the nematode. J Gerontol 48: B57-61.
  91. Fujita H, Ishii N, Suzuki K (1984) Effects of 8-methoxypsoralen plus near-ultraviolet light on the nematode Caenorhabditis elegans. Photochem Photobiol 39: 831-834.
  92. Bardin PG, van Eeden SF, Moolman JA, Foden AP, Joubert JR (1994) Organophosphate and carbamate poisoning. Arch Intern Med 154: 1433-1441.
  93. Kim Y, Sun H (2007) Functional genomic approach to identify novel genes involved in the regulation of oxidative stress resistance and animal lifespan. Aging Cell 6: 489-503
  94. Uversky VN (2004) Neurotoxicant-induced animal models of Parkinson's disease: understanding the role of rotenone, maneb and paraquat in neurodegeneration. Cell Tissue Res 318: 225-241.
  95. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, et al. (2000) Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci 3: 1301-1306.
  96. Gao HM, Liu B, Hong JS (2003) Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci 23: 6181-6187.
  97. Ishiguro H, Yasuda K, Ishii N, Ihara K, Ohkubo T, et al. (2001) Enhancement of oxidative damage to cultured cells and Caenorhabditis elegans by mitochondrial electron transport inhibitors. IUBMB life 51: 263-268.
  98. Wolozin B, Saha S, Guillily M, Ferree A, Riley M (2008) Investigating convergent actions of genes linked to familial Parkinson's disease. Neurodegener Dis 5: 182-185.
  99. Williams PL, Dusenbery DB (1990) A promising indicator of neurobehavioral toxicity using the nematode Caenorhabditis elegans and computer tracking. Toxicol Ind Health 6: 425-440.
  100. Williams PL, Dusenbery DB (1990) Aquatic toxicity testing using the nematode, Caenorhabditis elegans. Environ Toxicol Chem 9: 1285-1290.
  101. Anderson GL, Cole RD, Williams PL (2004) Assessing behavioral toxicity with Caenorhabditis elegans. Environ Toxicol Chem 23: 1235-1240.
  102. Melstrom PC, Williams PL (2007) Reversible AChE inhibitors in C. elegans vs. rats, mice. Biochem Biophys Res Commun 357: 200-205.
  103. Rajini PS, Melstrom PC, Williams PL (2008) A comparative study on the relationship between various toxicological endpoints in Caenorhabditis elegans exposed to organophosphorus insecticides. Toxicol Environ Health 71: 1043-1050.
  104. Saffih-Hdadi K, Bruckler L, Amichot M, Belzunces L (2005) Modeling impact of parathion and its metabolite paraoxon on the nematode Caenorhabditis elegans in soil. Environ Toxicol Chem 24: 1387-1394.
  105. Bushnell DM, Martin ML (1999) Quality of life and Parkinson's disease: translation and validation of the US Parkinson's Disease Questionnaire (PDQ-39). Qual Life Res 8: 345-350.
  106. Wilson JM, Levey AI, Rajput A, Ang L, Guttman M, et al. (1996) Differential changes in neurochemical markers of striatal dopamine nerve terminals in idiopathic Parkinson's disease. Neurology 47: 718-726.
  107. Dauer W, Przedborski S (2003) Parkinson's disease: mechanisms and models. Neuron 39: 889-909.
  108. Wood-Kaczmar A, Gandhi S, Wood NW (2006) Understanding the molecular causes of Parkinson's disease. Trends Mol Med 12: 521-528.
  109. Benedetto A, Au C, Aschner M, Nass R. Nass R, et al. (2008) Manganese and C. elegans in Parkinson's disease, Parkinson's Disease: Pathogenic and Therapeutic Insights from Toxin and Genetic Models.
  110. Ichibangase T, Saimaru H, Takamura N, Kuwahara T, Koyama A, et al. (2008) Proteomics of Caenorhabditis elegans over-expressing human alpha-synuclein analyzed by fluorogenic derivatization-liquid chromatography/tandem mass spectrometry: Identification of actin and several ribosomal proteins as negative markers at early Parkinson's disease stages. Biomed Chromatogr 22: 232-234.
  111. Lakso M, Vartiainen S, Moilanen AM, Sirvio J, Thomas JH, et al. (2003) Dopaminergic neuronal loss and motor deficits in Caenorhabditis elegans overexpressing human alpha-synuclein. J Neurochem 86: 165-172
  112. Springer W, Hoppe T, Schmidt E, Baumeister R (2005) A Caenorhabditis elegans Parkin mutant with altered solubility couples alpha-synuclein aggregation to proteotoxic stress. Hum Mol Genet 14: 3407-3423.
  113. Chiaki Fujitake MT, Inoue H, Takahashi K (2004) Characterization of C. elegans ubiquitin C-terminal hydrolases. In East Asia Worm Meeting 56: 20-26.
  114. Sakaguchi-Nakashima A, Meir JY, Jin Y, Matsumoto K, Hisamoto N (2007) LRK-1, a C. elegans PARK8-related kinase, regulates axonal-dendritic polarity of SV proteins. Curr Biol 17: 592-598.
  115. Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, et al. (2006) Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science 313: 324-328.
  116. Gitler AD, Bevis BJ, Shorter J, Strathearn KE, Hamamichi S, et al. (2008) The Parkinson's disease protein alpha-synuclein disrupts cellular Rab homeostasis. Proc Natl Acad Sci USA 105: 145-150.
  117. Hamamichi S, Rivas RN, Knight AL, Cao S, Caldwell KA, et al. (2008) Hypothesis-based RNAi screening identifies neuroprotective genes in a Parkinson's disease model. Proc Natl Acad Sci U S A 105: 728-733.
  118. van Ham TJ, Thijssen KL, Breitling R, Hofstra RM, Plasterk RH, Nollen EA (2008) C. elegans model identifies genetic modifiers of alpha-synuclein inclusion formation during aging. PLoS Genet 4: e1000027.
  119. Kopin IJ, Markey SP (1988) MPTP toxicity: implications for research in Parkinson's disease. Annu Rev Neurosci 11: 81-96.
  120. Langston JW, Langston EB, Irwin I (1984) MPTP-induced parkinsonism in human and non-human primates--clinical and experimental aspects. Acta Neurol Scand Suppl 100: 49-54.
  121. Cao S, Gelwix CC, Caldwell KA, Caldwell GA (2005) Torsin-mediated protection from cellular stress in the dopaminergic neurons of Caenorhabditis elegans. J Neurosci 25: 3801-3812.
  122. Marvanova M, Nichols CD (2007) Identification of neuroprotective compounds of Caenorhabditis elegans dopaminergic neurons against 6-OHDA. J Mol Neurosci 31: 127-137
  123. Nass R, Hall DH, Miller DM 3rd, Blakely RD (2002) Neurotoxin-induced degeneration of dopamine neurons in Caenorhabditis elegans. Proc Natl Acad Sci U S A 99: 3264-3269.
  124. Schmidt E, Seifert M, Baumeister R (2007) Caenorhabditis elegans as a model system for Parkinson's disease. Neurodegener Dis 4: 199-217.
  125. Brandt R, Gergou A, Wacker I, Fath T, Hutter HA (2007) Caenorhabditis elegans model of tau hyperphosphorylation: Induction of developmental defects by transgenic overexpression of Alzheimer's disease-like modified tau. Neurobiol Aging 30: 22-33.
  126. Dickey CA, Yue M, Lin WL, Dickson DW, Dunmore JH, et al. (2006) Deletion of the ubiquitin ligase CHIP leads to the accumulation, but not the aggregation, of both endogenous phospho- and caspase-3-cleaved tau species. J Neurosci 26: 6985-6996.
  127. Link CD (2001) Transgenic invertebrate models of age-associated neurodegenerative diseases. Mech Ageing Dev 122: 1639-1649.
  128. Kraemer BC, Burgess JK, Chen JH, Thomas JH, Schellenberg GD (2006) Molecular pathways that influence human tau-induced pathology in Caenorhabditis elegans. Hum Mol Genet 15: 1483-1496
  129. Kraemer BC, Schellenberg GD (2007) SUT-1 enables tau-induced neurotoxicity in C. elegans. Hum Mol Genet 16: 1959-1971.
  130. Baumeister R, Leimer U, Zweckbronner I, Jakubek C, Grunberg J, et al. (1997) Human presenilin-1, but not familial Alzheimer's disease (FAD) mutants, facilitate Caenorhabditis elegans Notch signalling independently of proteolytic processing. Genes Funct 149-159.
  131. Levitan D, Greenwald I (1995) Facilitation of lin-12-mediated signalling by sel-12, a Caenorhabditis elegans S182 Alzheimer's disease gene. Nature 377: 351-354
  132. Li X, Greenwald I (1997) HOP-1, a Caenorhabditis elegans presenilin, appears to be functionally redundant with SEL-12 presenilin and to facilitate LIN-12 and GLP-1 signaling. Proc Natl Acad Sci USA 94: 12204-12209.
  133. Kitagawa N, Shimohama S, Oeda T, Uemura K, Kohno R, et al. (2003) The role of the presenilin-1 homologue gene sel-12 of Caenorhabditis elegans in apoptotic activities. J Biol Chem 278: 12130-12134.
  134. Berezovska O, Frosch M, McLean P, Knowles R, Koo E, et al. (1999) The Alzheimer-related gene presenilin 1 facilitates notch 1 in primary mammalian neurons. Brain Res Mol Brain Res 69: 273-280.
  135. Ray WJ, Yao M, Nowotny P, Mumm J, Zhang W, et al. (1999) Evidence for a physical interaction between presenilin and Notch. Proc Natl Acad Sci U S A 96: 3263-3268.
  136. Niwa R, Zhou F, Li C, Slack FJ (2008) The expression of the Alzheimer's amyloid precursor protein-like gene is regulated by developmental timing microRNAs and their targets in Caenorhabditis elegans. Dev Biol 315: 418-425.
  137. Boyd-Kimball D, Poon HF, Lynn BC, Cai J, Pierce WMJr, et al. (2006) Proteomic identification of proteins specifically oxidized in Caenorhabditis elegans expressing human Abeta(1-42): Implications for Alzheimer's disease. Neurobiol Aging 27: 1239-1249.
  138. Drake J, Link CD, Butterfield DA (2003) Oxidative stress precedes fibrillar deposition of Alzheimer's disease amyloid beta-peptide (1-42) in a transgenic Caenorhabditis elegans model. Neurobiol Aging 24: 415-420.
  139. Gutierrez-Zepeda A, Luo Y (2004) Testing the amyloid toxicity hypothesis of Alzheimer's disease in transgenic Caenorhabditis elegans model Front. Biosci 9: 3333-3338.
  140. Wu Y, Luo Y (2005) Transgenic C. elegans as a model in Alzheimer's research. Curr Alzheimer Res 2: 37-45.
  141. Wu Y, Wu Z, Butko P, Christen Y, Lambert MP, et al. (2006) Amyloid-beta-induced pathological behaviors are suppressed by Ginkgo biloba extract EGb 761 and ginkgolides in transgenic Caenorhabditis elegans. J Neurosci 26: 13102-13113.
  142. Kraemer BC, Zhang B, Leverenz JB, Thomas JH, Trojanowski JQ, et al. (2003) Neurodegeneration and defective neurotransmission in a Caenorhabditis elegans model of tauopathy. Proc Natl Acad Sci 100: 9980-9985.
  143. Francis R, McGrath G, Zhang J, Ruddy DA, Sym M, et al. (2002) aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 3: 85-97.
  144. Link CD, Taft A, Kapulkin V, Duke K, Kim S, et al. (2003) Gene expression analysis in a transgenic Caenorhabditis elegans Alzheimer's disease model. Neurobiol Aging 24: 397-413.
  145. Florez-McClure ML, Hohsfield LA, Fonte G, Bealor MT, Link CD (2007) Decreased insulin-receptor signaling promotes the autophagic degradation of beta-amyloid peptide in C. elegans. Autophagy 3: 569-580.
  146. Fonte V, Kipp DR, Yerg JIII, Merin D, Forrestal M, et al. (2008) Suppression of in vivo beta-amyloid peptide toxicity by overexpression of the HSP-16.2 small chaperone protein. J Biol Chem 283: 784-791.
  147. Gutierrez-Zepeda A, Santell R, Wu Z, Brown M, Wu Y, et al. (2005) Soy isoflavone glycitein protects against beta amyloid-induced toxicity and oxidative stress in transgenic Caenorhabditis elegans. BMC Neurosci 6: 54.
  148. Luo Y (2006) Alzheimer's disease, the nematode Caenorhabditis elegans, and ginkgo biloba leaf extract. Life Sci 78: 2066-2072.
  149. Chopra VS, Metzler M, Rasper DM, Engqvist-Goldstein AE, Singaraja R, et al. (2000) HIP12 is a non-proapoptotic member of a gene family including HIP1, an interacting protein with huntingtin. Mamm Genome 11: 1006-1015.
  150. Holbert S, Dedeoglu A, Humbert S, Saudou F, Ferrante RJ, et al. (2003) Cdc42-interacting protein 4 binds to huntingtin: neuropathologic and biological evidence for a role in Huntington's disease. Proc Natl Acad Sci U S A 100: 2712-2717.
  151. Bates EA, Victor M, Jones AK, Shi Y, Hart AC (2006) Differential contributions of Caenorhabditis elegans histone deacetylases to huntingtin polyglutamine toxicity. J Neurosci 26: 2830-2838.
  152. Faber PW, Voisine C, King DC, Bates EA, Hart AC (2002) Glutamine/proline-rich PQE-1 proteins protect Caenorhabditis elegans neurons from huntingtin polyglutamine neurotoxicity. Proc Natl Acad Sci USA 99: 17131-17136.
  153. Anderson JL, Head SI, Rae C, Morley JW (2002) Brain function in Duchenne muscular dystrophy. Brain 125: 4-13.
  154. Blake DJ, Kröger S (2000) The neurobiology of duchenne muscular dystrophy: learning lessons from muscle? Trends Neurosci 23: 92-99.
  155. Poysky J (2007) Behavior patterns in Duchenne muscular dystrophy: Report on the Parent Project Muscular Dystrophy Behavior Workshop 8–9 of December 2006, Philadelphia, USA. Neuromuscul Disord 17: 986-994.
  156. Bessou C, Giugia JB, Franks CJ, Holden-Dye L, Segalat L (1998) Mutations in the Caenorhabditis elegans dystrophin-like gene dys-1 lead to hyperactivity and suggest a link with cholinergic transmission. Neurogenetics 61-72.
  157. Gieseler K, Bessou C, Segalat L (1999) Dystrobrevin- and dystrophin-like mutants display similar phenotypes in the nematode Caenorhabditis elegans. Neurogenetics 2: 87-90.
  158. Gieseler K, Abdel-Dayem M, Ségalat L (1999) In vitro interactions of Caenorhabditis elegans dystrophin with dystrobrevin and syntrophin. FEBS Lett 461: 59-62.
  159. Gieseler K, Grisoni K, Ségalat L (2000) Genetic suppression of phenotypes arising from mutations in dystrophin-related genes in Caenorhabditis elegans. Curr Biol 10: 1092-1097.
  160. Grisoni K, Martin E, Gieseler K, Mariol MC, Ségalat L (2002) Genetic evidence for a dystrophin-glycoprotein complex (DGC) in Caenorhabditis elegans. Gene 294: 77-86.
  161. Grisoni K, Gieseler K, Ségalat L (2002) Dystrobrevin requires a dystrophin-binding domain to function in Caenorhabditis elegans. Eur J Biochem 269: 1607-1612.
  162. Gieseler K, Grisoni K, Mariol MC, Segalat L (2002) Overexpression of dystrobrevin delays locomotion defects and muscle degeneration in a dystrophin-deficient Caenorhabditis elegans. Neuromuscul Disord 12: 371-377.
  163. Grisoni K, Gieseler K, Mariol MC, Martin E, Carre-Pierrat M, et al. (2003) The stn-1 syntrophin gene of C.elegans is functionally related to dystrophin and dystrobrevin. J Mol Biol 332: 1037-1046.
  164. Carre-Pierrat M, Mariol MC, Chambonnier L, Laugraud A, Heskia F, et al. (2006) Blocking of striated muscle degeneration by serotonin in C. elegans. J Muscle Res Cell Motil 27: 253-258.
  165. Gaud A, Simon JM, Witzel T, Carre-Pierrat M, Wermuth CG, et al. (2004) Prednisone reduces muscle degeneration in dystrophin-deficient Caenorhabditis elegans. Neuromuscul Disord 14: 365-370.
  166. Nyamsuren O, Faggionato D, Loch W, Schulze E, Baumeister R (2007) A mutation in CHN-1/CHIP suppresses muscle degeneration in Caenorhabditis elegans. Dev Biol 312: 193-202.
  167. Boulton SJ, Gartner A, Reboul J, Vaglio P, Dyson N, et al. (2002) Combined functional genomic maps of the C. elegans DNA damage response. Science 295: 127-131.
  168. Hevelone J, Hartman PS (1988) An endonuclease from Caenorhabditis elegans: partial purification and characterization. Biochem Genet 26: 447-461.
  169. O'Neil N, Rose A (2005) DNA repair, WormBook. , 2005 The C. elegans Research Community.
  170. Stergiou L, Hengartner MO (2004) Death and more: DNA damage response pathways in the nematode C. elegans. Cell Death Differ 11: 21-28.
  171. Hartman PS (1985) Epistatic interactions of radiation-sensitive (rad) mutants of Caenorhabditis elegans. Genetics 109: 81-93.
  172. Hartman PS, Herman RK (1982) Radiation-sensitive mutants of Caenorhabditis elegans. Genetics 102: 159-178.
  173. Hyun M, Lee J, Lee K, May A, Bohr VA, et al. (2008) Longevity and resistance to stress correlate with DNA repair capacity in Caenorhabditis elegans. Nucleic Acids Res 36: 1380-1389.
  174. Meyer JN, Boyd WA, Azzam GA, Haugen AC, Freedman JH, et al. (2007) Decline of nucleotide excision repair capacity in aging Caenorhabditis elegans. Genome Biol 8: R70.
  175. Friedberg EC, Walker GC, Siede W, Wood RD, Schultz RA, et al. (2006) DNA Repair and Mutagenesis. ASM.
  176. Truglio JJ, Croteau DL, Van Houten B, Kisker C (2006) Prokaryotic nucleotide excision repair: the UvrABC system. Chem Rev 106: 233-252.
  177. Boulton SJ, Martin JS, Polanowska J, Hill DE, Gartner A, et al. (2004) BRCA1/BARD1 orthologs required for DNA repair in Caenorhabditis elegans. Curr Biol 14: 33-39.
  178. van Haaften G, Vastenhouw NL, Nollen EA, Plasterk RH, Tijsterman M (2004) Gene interactions in the DNA damage-response pathway identified by genome-wide RNA-interference analysis of synthetic lethality. Proc Natl Acad Sci U S A 101: 12992-12996.
  179. Dequen F, Gagnon SN, Desnoyers S (2005) Ionizing radiations in Caenorhabditis elegans induce poly(ADP-ribosyl)ation, a conserved DNA-damage response essential for survival. DNA Repair (Amst) 4: 814-825.
  180. Gagnon SN, Hengartner MO, Desnoyers S (2002) The genes pme-1 and pme-2 encode two poly(ADP-ribose) polymerases in Caenorhabditis elegans. Biochem J 368: 263-271.
  181. Hartman PS, Hevelone J, Dwarakanath V, Mitchell DL (1989) Excision repair of UV radiation-induced DNA damage in Caenorhabditis elegans. Genetics 122: 379-385.
  182. Kanugula S, Pegg AE (2001) Novel DNA repair alkyltransferase from Caenorhabditis elegans. Environ Mol Mutagen 38: 235-243.
  183. Munakata N, Morohoshi F (1986) DNA glycosylase activities in the nematode Caenorhabditis elegans. Mutat Res 165: 101-107.
  184. Shatilla A, Leduc A, Yang XM, Ramotar D (2005) Identification of two apurinic/apyrimidinic endonucleases from Caenorhabditis elegans by cross-species complementation. DNA Repair 4: 655-670.
  185. Shatilla A, Ramotar D (2002) Embryonic extracts derived from the nematode Caenorhabditis elegans remove uracil from DNA by the sequential action of uracil-DNA glycosylase and AP (apurinic/apyrimidinic) endonuclease. Biochem J 365: 547-553.
  186. Astin JW, O'Neil NJ, Kuwabara PE (2008) Nucleotide excision repair and the degradation of RNA pol II by the Caenorhabditis elegans XPA and Rsp5 orthologues, RAD-3 and WWP-1. DNA Repair 7: 267-280.
  187. Dequen F, St-Laurent JF, Gagnon SN, Carreau M, Desnoyers S (2005) The Caenorhabditis elegans FancD2 ortholog is required for survival following DNA damage. Comp Biochem Physiol B Biochem Mol Biol 141: 453-460.
  188. Lee MH, Ahn B, Choi IS, Koo HS (2002) The gene expression and deficiency phenotypes of Cockayne syndrome B protein in Caenorhabditis elegans. FEBS Lett 522: 47-51.
  189. Park HK, Suh D, Hyun M, Koo HS, Ahn B (2004) A DNA repair gene of Caenorhabditis elegans: a homolog of human XPF. DNA Repair (Amst) 3: 1375-1383.
  190. Park HK, Yook JS, Koo HS, Choi IS, Ahn B (2002) The Caenorhabditis elegans XPA homolog of human XPA. Mol Cells 14: 50-55.
  191. St-Laurent JF, Gagnon SN, Dequen F, Hardy I, Desnoyers S (2007) Altered DNA damage response in Caenorhabditis elegans with impaired poly(ADP-ribose) glycohydrolases genes expression. DNA Repair (Amst) 6: 329-343.
  192. Aoki H, Sato S, Takanami T, Ishihara T, Katsura I, et al. (2000) Characterization of Ce-atl-1, an ATM-like gene from Caenorhabditis elegans. Mol Gen Genet 264: 119-126.
  193. Kelly KO, Dernburg AF, Stanfield GM, Villeneuve AM (2000) Caenorhabditis elegans msh-5 is required for both normal and radiation-induced meiotic crossing over but not for completion of meiosis. Genetics 156: 617-630
  194. Sadaie T, Sadaie Y (1989) Rad-2-dependent repair of radiation-induced chromosomal-aberrations in Caenorhabditis-elegans. Mutat Res 218: 25-31.
  195. Takanami T, Sato S, Ishihara T, Katsura I, Takahashi H, et al. (1998) Characterization of a Caenorhabditis elegans recA-like gene Ce-rdh-1 involved in meiotic recombination. DNA Res 5: 373-377.
  196. Ahmed S, Alpi A, Hengartner MO, Gartner A (2001) C. elegans RAD-5/CLK-2 defines a new DNA damage checkpoint protein. Curr Biol 11: 1934-1944.
  197. Ahmed S, Hodgkin J (2000) MRT-2 checkpoint protein is required for germline immortality and telomere replication in C. elegans. Nature 403: 159-164.
  198. Gartner A, Milstein S, Ahmed S, Hodgkin J, Hengartner MO (2000) A conserved checkpoint pathway mediates DNA damage--induced apoptosis and cell cycle arrest in C. elegans. Mol Cell 5: 435-443.
  199. Jagasia R, Grote P, Westermann B, Conradt B (2005) DRP-1-mediated mitochondrial fragmentation during EGL-1-induced cell death in C. elegans. Nature 433: 754-760.
  200. Kinchen JM, Hengartner MO (2005) Tales of cannibalism, suicide, and murder: Programmed cell death in C. elegans. Curr Top Dev Biol 65: 1-45.
  201. Lettre G, Hengartner MO (2006) Developmental apoptosis in C. elegans: a complex CEDnario. Nat Rev Mol Cell Biol 7: 97-108.
  202. Olsen A, Vantipalli MC, Lithgow GJ (2006) Using Caenorhabditis elegans as a model for aging and age-related diseases. Ann N Y Acad Sci 1067: 120-128.
  203. Schumacher B, Schertel C, Wittenburg N, Tuck S, Mitani S, et al. (2005) C. elegans ced-13 can promote apoptosis and is induced in response to DNA damage. Cell Death Differ 12: 153-161.
  204. Stergiou L, Doukoumetzidis K, Sendoel A, Hengartner MO (2007) The nucleotide excision repair pathway is required for UV-C-induced apoptosis in Caenorhabditis elegans. Cell Death Differ 14: 1129-1138.
  205. Ahmed S (2006) Uncoupling of pathways that promote postmitotic life span and apoptosis from replicative immortality of Caenorhabditis elegans germ cells. Aging Cell 5: 559-563.
  206. Clejan I, Boerckel J, Ahmed S (2006) Developmental modulation of nonhomologous end joining in Caenorhabditis elegans. Genetics 173: 1301-1317.
  207. Pinkston-Gosse J, Kenyon C (2007) DAF-16/FOXO targets genes that regulate tumor growth in Caenorhabditis elegans. Nat Genet 39: 1403-1409.
  208. Poulin G, Nandakumar R, Ahringer J (2004) Genome-wide RNAi screens in Caenorhabditis elegans: impact on cancer research. Oncogene 23: 8340-8345.
  209. Mee CJ, Tomlinson SR, Perestenko PV, De Pomerai D, Duce IR, et al. (2004) Latrophilin is required for toxicity of black widow spider venom in Caenorhabditis elegans. Biochem J 378: 185-191.
  210. van Haaften G, Plasterk RH, Tijsterman M (2004) Genomic instability and cancer: scanning the Caenorhabditis elegans genome for tumor suppressors. Oncogene 23: 8366-8375.
  211. Antebi A (2007) Genetics of aging in Caenorhabditis elegans. PLoS Genet 3: 1565-1571.
  212. Brys K, Vanfleteren JR, Braeckman BP (2007) Testing the rate-of-living/oxidative damage theory of aging in the nematode model Caenorhabditis elegans. Exp Gerontol 42: 845-851.
  213. Johnson TE (2003) Advantages and disadvantages of Caenorhabditis elegans for aging research. Exp Gerontol 38: 1329-1332.
  214. Kenyon C (2005) The plasticity of aging: insights from long-lived mutants. Cell 120: 449-460.
  215. Rea SL, Ventura N, Johnson TE (2007) Relationship between mitochondrial electron transport chain dysfunction, development, and life extension in Caenorhabditis elegans. PLoS Biol 5: e259.
  216. Ventura N, Rea SL, Testi R (2006) Long-lived C. elegans mitochondrial mutants as a model for human mitochondrial-associated diseases. Exp Gerontol 41: 974-991.
  217. Barrett PL, Fleming JT, Göbel V (2004) Targeted gene alteration in Caenorhabditis elegans by gene conversion. Nat Genet 36: 1231-1237.
  218. Berezikov E, Bargmann CI, Plasterk RH (2004) Homologous gene targeting in Caenorhabditis elegans by biolistic transformation. Nucleic Acids Res 32: e40.
  219. Plasterk RHA, Groenen JTM (1992) Targeted alterations of the Caenorhabditis-elegans genome by transgene instructed DNA double strand break repair following Tc1 excision. EMBO J 11: 287-290.
  220. Rushforth AM, Saari B, Anderson P (1993) Site-selected insertion of the transposon Tc1 into a Caenorhabditis elegans myosin light chain gene. Mol Cell Biol 13: 902-910.
  221. Coohill T, Marshall T, Schubert W, Nelson G (1988) Ultraviolet mutagenesis of radiation-sensitive (rad) mutants of the nematode Caenorhabditis elegans. Mutat Res 209: 99-106.
  222. Hartman PS (1984) UV irradiation of wild-type and radiation-sensitive mutants of the nematode Caenorhabditis elegans—Fertilities, survival, and parental effects, Photochem. Photobiol 39: 169-175.
  223. Jones CA, Hartman PS (1996) Replication in UV-irradiated Caenorhabditis elegans embryos. Photochem Photobiol 63: 187-192.
  224. Keller C, Calkins J, Hartman PS, Rupert CS. UV photobiology of the nematode Caenorhabditis-elegans—Action spectra, absence of photoreactivation and effects of caffeine. Photochem Photobiol 46: 488.
  225. Stewart H, Rosenbluth RE, Baillie DL (1991) Most ultraviolet-irradiation induced mutations in the nematode Caenorhabditis-elegans are chromosomal rearrangements. Mutat Res 249: 37-54.
  226. Weidhaas JB, Eisenmann DM, Holub JM, Nallur SV (2006) A Caenorhabditis elegans tissue model of radiation-induced reproductive cell death. Proc Natl Acad Sci U S A 103: 9946-9951.
  227. Neher DA, Sturzenbaum SR (2006) Extra-long PCR, an identifier of DNA adducts in single nematodes (Caenorhabditis elegans). Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 144: 279-285.
  228. Menzel R, Yeo HL, Rienau S, Li S, Steinberg CE, et al. (2007) Cytochrome P450s and short-chain dehydrogenases mediate the toxicogenomic response of PCB52 in the nematode Caenorhabditis elegans. J Mol Biol 370: 1-13.
  229. Holway AH, Kim SH, La Volpe A, Michael WM (2006) Checkpoint silencing during the DNA damage response in Caenorhabditis elegans embryos. J Cell Biol 172: 999-1008.
  230. Hartman PS, Marshall A (1992) Inactivation of wild-type and rad mutant Caenorhabditis elegans by 8-methoxypsoralen and near ultraviolet radiation. Photochem. Photobiol 55: 103-111.
  231. Mills DK, Hartman PS (1998) Lethal consequences of simulated solar radiation on the nematode Caenorhabditis elegans in the presence and absence of photosensitizers. Photochem. Photobiol 68: 816-823
  232. Hartman P, Ponder R, Lo HH, Ishii N (2004) Mitochondrial oxidative stress can lead to nuclear hypermutability. Mech Ageing Dev 125: 417-420.
  233. Salinas LS, Maldonado E, Navarro RE (2006) Stress-induced germ cell apoptosis by a p53 independent pathway in Caenorhabditis elegans. Cell Death Differ 13: 2129-2139.
  234. Ishi N, Suzuki N, Hartman PS, Suzuki K (1993) The radiation-sensitive mutant rad-8 of Caenorhabditis elegans is hypersensitive to the effects of oxygen on aging and development. Mech Ageing Dev 68: 1-10.
  235. Gibb S (2008) Toxicity testing in the 21st century: A vision and a strategy. Reprod Toxicol 25: 136-138.
  236. Dix DJ, Houck KA, Martin MT, Richard AM, Setzer RW, et al. (2007) The ToxCast program for prioritizing toxicity testing of environmental chemicals. Toxicol Sci 95: 5-12.
  237. Baek JH, Cosman P, Feng Z, Silver J, Schafer WR (2002) Using machine vision to analyze and classify Caenorhabditis elegans behavioral phenotypes quantitatively. J Neurosci Methods 118: 9-21.
  238. Bennett JL, Pax RA (1986) Micromotility meter: An instrument designed to evaluate the action of drugs on motility of larval and adult nematodes. Parasitology 93: 341-346.
  239. Hoshi K, Shingai R (2006) Computer-driven automatic identification of locomotion states in Caenorhabditis elegans. J Neurosci Methods 157: 355-363.
  240. Simonetta SH, Golombek DA (2007) An automated tracking system for Caenorhabditis elegans locomotor behavior and circadian studies application. J Neurosci Methods 161: 273-280.
  241. Tsibidis GD, Tavernarakis N (2007) Nemo: a computational tool for analyzing nematode locomotion. BMC Neurosci 8: 86.
  242. Black MC, Williams PL (2001) Preliminary assessment of metal toxicity in the middle Tisza River (Hungary) flood plain. J Soils Sediments 1: 203-206.
  243. Burns AR, Kwok TC, Howard A, Houston E, Johanson K, et al. (2006) High-throughput screening of small molecules for bioactivity and target identification in Caenorhabditis elegans. Nat Protoc 1: 1906-1914.
  244. Dupuy D, Bertin N, Hidalgo CA, Venkatesan K, Tu D, et al. (2007) Genome-scale analysis of in vivo spatiotemporal promoter activity in Caenorhabditis elegans. Nat Biotechnol 25: 663-668.
  245. Rohde CB, Zeng F, Gonzalez-Rubio R, Angel M, Yanik MF (2007) Microfluidic system for on-chip high-throughput whole-animal sorting and screening at subcellular resolution. Proc Natl Acad Sci USA 104: 13891-13895.
  246. Miller LM, Hartman PS (1998) The effects of benzo[a]pyrene (cough cough!) on C. elegans. Worm Breed Gaz 15: 43.
  247. Griffitts JS, Haslam SM, Yang T, Garczynski SF, Mulloy B, et al. (2005) Glycolipids as receptors for Bacillus thuringiensis crystal toxin. Science 307: 922-925.
  248. Griffitts JS, Whitacre JL, Stevens DE, Aroian RV (2001) Bt toxin resistance from loss of a putative carbohydrate-modifying enzyme. Science 293: 860-864.
  249. Harris TW, Chen N, Cunningham F, Tello-Ruiz M, Antoshechkin, et al. (2004) WormBase: a multi-species resource for nematode biology and genomics. Nucleic Acids Res 32: D411-417.
  250. Kwon JY, Hong M, Choi MS, Kang SJ, Duke K, et al. (2004) Ethanol-response genes and their regulation analyzed by a microarray and comparative genomic approach in the nematode Caenorhabditis elegans. Genomics 83: 600-614
  251. Custodia N, Won SJ, Novillo A, Wieland M, Li C, et al. (2001) Caenorhabditis elegans as an environmental monitor using DNA microarray analysis, Environmental Hormones: the Scientific Basis of Endocrine Disruption. Ann N Y Acad Sci 948: 32-42.
  252. Reichert K, Menzel R (2005) Expression profiling of five different xenobiotics using a Caenorhabditis elegans whole genome microarray. Chemosphere 61: 229-237.
  253. Simmer F, Tijsterman M, Parrish S, Koushika SP, Nonet ML, et al. (2002) Loss of the putative RNA-directed RNA polymerase RRF-3 makes C. elegans hypersensitive to RNAi. Curr Biol 12: 1317-1319.
  254. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, et al. (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391: 806-811.
  255. Fewell GD, Schmitt K (2006) Vector-based RNAi approaches for stable, inducible and genome-wide screens. Drug Discov Today 11: 975-982.
  256. Kamath RS, Ahringer J (2003) Genome-wide RNAi screening in Caenorhabditis elegans. Methods 30: 313-321.
  257. Simmer F, Moorman C, van der Linden AM, Kuijk E, van den Berghe PV, et al. (2003) Genome-wide RNAi of C. elegans using the hypersensitive rrf-3 strain reveals novel gene functions. PLoS Biol 1: E12.
  258. Esposito G, Di Schiavi E, Bergamasco C, Bazzicalupo P (2007) Efficient and cell specific knock-down of gene function in targeted C. elegans neurons. Gene 395: 170-176.
  259. Lee RC, Hammell CM, Ambros V (2006) Interacting endogenous and exogenous RNAi pathways in Caenorhabditis elegans. RNA 12: 589-597.
  260. Tops BB, Tabara H, Sijen T, Simmer F, Mello CC, et al. (2005) RDE-2 interacts with MUT-7 to mediate RNA interference in Caenorhabditis elegans. Nucleic Acids Res 33: 347-355.
  261. Tabara H, Yigit E, Siomi H, Mello CC (2002) The dsRNA binding protein RDE-4 interacts with RDE-1, DCR-1, and a DExH-box helicase to direct RNAi in C. elegans. Cell 109: 861-871.
  262. Ayyadevara S, Dandapat A, Singh SP, Siegel ER, Shmookler Reis RJ, et al. (2007) Life span and stress resistance of Caenorhabditis elegans are differentially affected by glutathione transferases metabolizing 4-hydroxynon-2-enal. Mech Ageing Dev 128: 196-205.
  263. Ichishita R, Tanaka K, Sugiura Y, Sayano T, Mihara K, et al. (2008) An RNAi screen for mitochondrial proteins required to maintain the morphology of the organelle in Caenorhabditis elegans. J Biochem 143: 449-454.
  264. Lee SS, Lee RY, Fraser AG, Kamath RS, Ahringer J, et al. (2003) A systematic RNAi screen identifies a critical role for mitochondria in C. elegans longevity. Nat Genet 33: 40-48.
  265. Schmitz C, Kinge P, Hutter H (2007) Axon guidance genes identified in a large-scale RNAi screen using the RNAi-hypersensitive Caenorhabditis elegans strain nre-1(hd20) lin-15b(hd126). Proc Natl Acad Sci USA 104: 834-839.
  266. Zhang SL, Yeromin AV, Zhang XH, Yu Y, Safrina O, et al. (2006) Genome-wide RNAi screen of Ca(2+) influx identifies genes that regulate Ca(2+) release-activated Ca(2+) channel activity. Proc Natl Acad Sci USA 103: 9357-9362.
  267. Dhawan R, Dusenbery DB, Williams PL (1999) Comparison of lethality, reproduction, and behavior as toxicological endpoints in the nematode Caenorhabditis elegans. J Toxicol Environ Health A 58: 451-462.
  268. Cole RD, Anderson GL, Williams PL (2004) The nematode Caenorhabditis elegans as a model of organophosphate-induced mammalian neurotoxicity. Toxicol Appl Pharmacol 194: 248-256.
  269. Tatara CP, Newman MC, McCloskey JT, Williams PL (1998) Use of ion characteristics to predict relative toxicity of mono-, di- and trivalent metal ions: Caenorhabditis elegans LC50. Aquat Toxicol 42: 255-269.
  270. Peredney CL, Williams PL (2000) Utility of Caenorhabditis elegans for assessing heavy metal contamination in artificial soil. Arch Environ Contam Toxicol 39: 113-118.
  271. Peterson RT, Nass R, Boyd WA, Freedman JH, Dong K, et al. (2008) Use of non-mammalian alternative models for neurotoxicological study. Neurotoxicology 29: 546-555.
  272. Dengg M, van Meel JC (2004) Caenorhabditis elegans as model system for rapid toxicity assessment of pharmaceutical compounds. J Pharmacol Toxicol Methods 50: 209-214.
  273. Cioci LK, Qiu L, Freedman JH (2000) Transgenic strains of the nematode Caenorhabditis elegans as biomonitors of metal contamination. Environ Toxicol Chem 19: 2122-2129.
  274. Hasegawa K, Miwa S, Isomura K, Tsutsumiuchi K, Taniguchi H, et al. (2008) Acrylamide-responsive genes in the nematode Caenorhabditis elegans. Toxicol Sci 101: 215-225.
  275. Denver DR, Feinberg S, Steding C, Durbin M, Lynch M (2006) The relative roles of three DNA repair pathways in preventing Caenorhabditis elegan mutation accumulation. Genetics 174: 57-65.
  276. Jones D, Stringham EG, Babich SL, Candido EP (1996) Transgenic strains of the nematode C. elegans in biomonitoring and toxicology: Effects of captan and related compounds on the stress response. Toxicology 109: 119-127.
  277. Chu KW, Chan SK, Chow KL (2005) Improvement of heavy metal stress and toxicity assays by coupling a transgenic reporter in a mutant nematode strain. Aquat Toxicol 74: 320-332.
  278. Easton A, Guven K, de Pomerai DI (2001) Toxicity of the dithiocarbamate fungicide mancozeb to the nontarget soil nematode, Caenorhabditis elegans. J Biochem Mol Toxicol 15: 15-25.
  279. Mutwakil MH, Reader JP, Holdich DM, Smithurst PR, Candido EPM, et al. (1997) Use of stress-inducible transgenic nematodes as biomarkers of heavy metal pollution in water samples from an English river system. Arch Environ Contam Toxicol 32: 146-153.
  280. Roh JY, Lee J, Choi J (2006) Assessment of stress-related gene expression in the heavy metal-exposed nematode Caenorhabditis elegans: A potential biomarker for metal-induced toxicity monitoring and environmental risk assessment. Environ Toxicol Chem 25: 2946-2956.
  281. Höss S, Weltje L (2007) Endocrine disruption in nematodes: effects and mechanisms. Ecotoxicology 16: 15-28.
  282. Freedman JH, Chen M, Coughlan S, Boyd WA (2006) Changes in Gene Expression due to Chronic Exposure to Environmental Pollutants. Aquat Toxicol 90: 161-171.
  283. Kammenga JE, Dallinger R, Donker MH, Köhler HR, Simonsen V, et al. (2000) Biomarkers in terrestrial invertebrates for ecotoxicological soil risk assessment. Rev Environ Contam Toxicol 164: 93-147.
  284. Donkin SG, Dusenberry DB (1993) A soil toxicity test using the nematode Caenorhabditis elegans and an effective method of recovery. Arch Environ Contam Toxicol 25: 145-151.
  285. Graves AL, Boyd WA, Williams PL (2005) Using transgenic Caenorhabditis elegans in soil toxicity testing. Arch Environ Contam Toxicol 48: 490-494.
  286. American Society for Testing and Materials (ASTM) (2002) Standard guide for conducting laboratory soil toxicity tests with the nematode Caenorhabditis elegans. ASTM 11: 1606–1616.
  287. Barrows BD, Griffitts JS, Aroian RV (2006) Caenorhabditis elegans carbohydrates in bacterial toxin resistance. Methods Enzymol 417: 340-358.
  288. Barrows BD, Griffitts JS, Aroian RV (2007) Resistance is non-futile: resistance to Cry5B in the nematode Caenorhabditis elegans. J Invertebr Pathol 95: 198-200.
  289. Barrows BD, Haslam SM, Bischof LJ, Morris HR, Dell A, et al. (2007) Resistance to Bacillus thuringiensis toxin in Caenorhabditis elegans from loss of fucose. J Biol Chem 282: 3302-3311.
  290. Berezovska O, Xia MQ, Hyman BT (1998) Notch is expressed in adult brain, is coexpressed with presenilin-1, and is altered in Alzheimer disease. J Neuropathol Exp Neurol 57: 738-745.
  291. Bezchlebova J, Cernohlavkova J, Lana J, Sochova I, Kobeticova K, et al. (2007) Effects of toxaphene on soil organisms. Ecotoxicol Environ Saf 68: 326-334.
  292. Braungart E, Gerlach M, Riederer P, Baumeister R, Hoener MC (2004) Caenorhabditis elegans MPP+ model of Parkinson's disease for high-throughput drug screenings. Neurodegener Dis 1: 175-183.
  293. C elegans Sequencing Consortium (1998) Genome sequence of the nematode C. elegans: a platform for investigating biology. Science 282: 2012-2018.
  294. Cheng Q, Valmas N, Reilly PE, Collins PJ, Kopittke R, et al. (2003) Caenorhabditis elegans mutants resistant to phosphine toxicity show increased longevity and cross-resistance to the synergistic action of oxygen. Toxicol Sci 73: 60-65.
  295. Degtyareva NP, Greenwell P, Hofmann ER, Hengartner MO, Zhang L, et al. (2002) Caenorhabditis elegans DNA mismatch repair gene msh-2 is required for microsatellite stability and maintenance of genome integrity. Proc Natl Acad Sci USA 99: 2158-2163.
  296. Denver DR, Morris K, Lynch M, Thomas WK (2004) High mutation rate and predominance of insertions in the Caenorhabditis elegans nuclear genome. Nature 430: 679-682.
  297. Denver DR, Morris K, Lynch M, Vassilieva LL, Thomas WK (2000) High direct estimate of the mutation rate in the mitochondrial genome of Caenorhabditis elegans. Science 289: 2342-2344.
  298. Faber PW, Alter JR, MacDonald ME, Hart AC (1999) Polyglutamine-mediated dysfunction and apoptotic death of a Caenorhabditis elegans sensory neuron. Proc Natl Acad Sci USA 96: 179-184.
  299. Gieseler K, Mariol MC, Bessou C, Migaud M, Franks CJ, et al. (2001) Molecular, genetic and physiological characterisation of dystrobrevin-like (dyb-1) mutants of Caenorhabditis elegans. J Mol Biol 307: 107-117.
  300. Goldstein P, Modric T (1994) Transgenerational, ultrastructural analysis on the antioxidative effects of tocopherol on early gametogenesis in Caenorhabditis elegans grown in 100% oxygen. Toxicol Appl Pharmacol 124: 212-220.
  301. Greenwald IS, Horvitz HR (1980) unc-93(e1500): A behavioral mutant of Caenorhabditis elegans that defines a gene with a wild-type null phenotype. Genetics 96: 147-164.
  302. Griffitts JS, Huffman DL, Whitacre JL, Barrows BD, Marroquin LD, et al. Resistance to a bacterial toxin is mediated by removal of a conserved glycosylation pathway required for toxin-host interactions. J Biol Chem 278: 45594-45602.
  303. Guven K, Power RS, Avramides S, Allender R, de Pomerai D (1999) The toxicity of dithiocarbamate fungicides to soil nematodes, assessed using a stress-inducible transgenic strain of Caenorhabditis elegans. J Biochem Mol Toxicol 13: 324-333.
  304. Hamilton B, Dong Y, Shindo M, Liu W, Odell I, et al. (2005) A systematic RNAi screen for longevity genes in C. elegans. Genes Dev 19: 1544-1555.
  305. Hartman PS, Nelson GA, Nickoloff JA, Hoekstra MF (1998) Processing of DNA damage in the nematode Caenorhabditis elegans, DNA damage and Repair DNA Repair in Prokaryotes and Lower Eukaryotes. 1: 557-576.
  306. Hasegawa A, van der Bliek AM (2007) Inverse correlation between expression of the Wolfs Hirschhorn candidate gene Letm1 and mitochondrial volume in C. elegans and in mammalian cells. Hum Mol Genet 16: 2061-2071.
  307. Hasshoff M, Bohnisch C, Tonn D, Hasert B, Schulenburg H (2007) The role of Caenorhabditis elegans insulin-like signaling in the behavioral avoidance of pathogenic Bacillus thuringiensis. FASEB 21: 1801-1812.
  308. He L, He X, Lowe SW, Hannon GJ (2007) MicroRNAs join the p53 network--another piece in the tumour-suppression puzzle. Nat Rev Cancer 7: 819-822.
  309. Hitchcock DR, Black MC, Williams PL (1997) Investigations into using the nematode Caenorhabditis elegans for municipal and industrial wastewater toxicity testing. Arch. Environ. Contam. Toxicol. 33: 252-260.
  310. Höss S, Arndt M, Baumgarte S, Tebbe CC, Nguyen HT, et al. (2008) Effects of transgenic corn and Cry1Ab protein on the nematode, Caenorhabditis elegans. Ecotoxicol Environ Saf 70: 334-340.
  311. Hoss S, Haitzer M, Traunspurger W, Gratzer H, Ahlf W, et al. (1997) Influence of particle size distribution and content of organic matter on the toxicity of copper in sediment bioassays using Caenorhabditis elegans (nematoda). Water Air Soil Pollut 99: 689-695.
  312. Hoss S, Haitzer M, Traunspurger W, Steinberg CEW (1999) Growth and fertility of Caenorhabditis elegans (Nematoda) in unpolluted freshwater sediments: Response to particle size distribution and organic content. Environ Toxicol Chem 18: 2921-2925.
  313. Höss S, Henschel T, Haitzer M, Traunspurger W, Steinberg CE (2001) Toxicity of cadmium to Caenorhabditis elegans (Nematoda) in whole sediment and pore water--the ambiguous role of organic matter. Environ Toxicol Chem 20: 2794-2801.
  314. Huffman DL, Abrami L, Sasik R, Corbeil J, van der Goot FG, et al. (2002) Mitogen-activated protein kinase pathways defend against bacterial pore-forming toxins. Proc Natl Acad Sci USA 101: 10995-11000.
  315. Huffman DL, Bischof LJ, Griffitts JS, Aroian RV (2004) Pore worms: using Caenorhabditis elegans to study how bacterial toxins interact with their target host. Int J Med Microbiol 293: 599-607.
  316. Ibiam U, Grant A (2005) RNA/DNA ratios as a sublethal endpoint for large-scale toxicity tests with the nematode Caenorhabditis elegans. Environ. Toxicol Chem 24: 1155-1159.
  317. Jones D, Candido EP (1999) Feeding is inhibited by sublethal concentrations of toxicants and by heat stress in the nematode Caenorhabditis elegans: Relationship to the cellular stress response. J Exp Zool 284: 147-157.
  318. Kell A, Ventura N, Kahn N, Johnson TE (2007) Activation of SKN-1 by novel kinases in Caenorhabditis elegans. Free Radic Biol Med 43: 1560-1566.
  319. Klass M, Nguyen PN, Dechavigny A (1983) Age-correlated changes in the DNA template in the nematode Caenorhabditis elegans. Mech Ageing Dev 22: 253-263.
  320. Klass MR (1977) Aging in nematode Caenorhabditis elegans—Major biological and environmental factors influencing lifespan. Mech Ageing Dev 6: 413-429
  321. Krøll J (2007) Molecular chaperones and the epigenetics of longevity and cancer resistance. Ann N Y Acad Sci 1100: 75-83.
  322. Kurz CL, Shapira M, Chen K, Baillie DL, Tan MW (2007) Caenorhabditis elegans pgp-5 is involved in resistance to bacterial infection and heavy metal and its regulation requires TIR-1 and a p38 map kinase cascade. Biochem Biophys Res Commun 363: 438-443
  323. Kuwahara T, Koyama A, Gengyo-Ando K, Masuda M, Kowa H, et al. (2006) Familial Parkinson mutant alpha-synuclein causes dopamine neuron dysfunction in transgenic Caenorhabditis elegans. J Biol Chem 281: 334-340
  324. Lagido C, Pettitt J, Porter AJR, Paton GI, Glover LA (2001) Development and application of bioluminescent Caenorhabditis elegans as multicellular eukaryotic biosensors. FEBS Lett 493: 36-39
  325. Lee I, Lehner B, Crombie C, Wong W, Fraser AG, (2008) A single gene network accurately predicts phenotypic effects of gene perturbation in Caenorhabditis elegans. Nat Genet 40: 181-188
  326. Lee SJ, Yook JS, Han SM, Koo HS (2004) A Werner syndrome protein homolog affects C. elegans development, growth rate, life span and sensitivity to DNA damage by acting at a DNA damage checkpoint. Development 131: 2565-2575
  327. Li S, Armstrong CM, Bertin N, Ge H, Milstein S, et al. (2004) A map of the interactome network of the metazoan C. elegans. Science 303: 540-543.
  328. Marroquin LD, Elyassnia D, Griffitts JS, (2005) Bacillus thuringiensis (Bt) toxin susceptibility and isolation of resistance mutants in the nematode Caenorhabditis elegans. Genetics. 155: 1693-1699
  329. Murakami S (2007) Caenorhabditis elegans as a model system to study aging of learning and memory. Mol Neurobiol 35: 85-94.
  330. Murakami S, Johnson TE (1996) A genetic pathway conferring life extension and resistance to UV stress in Caenorhabditis elegans. Genetics 143: 1207-1218.
  331. National Research Council (NRC) (2000) Scientific Frontiers in Developmental Toxicology and Risk Assessment. DCThe National Academies Press 296-308.
  332. Parker JA, Connolly JB, Wellington C, Hayden M, Dausset J, et al. (2001) Expanded polyglutamines in Caenorhabditis elegans cause axonal abnormalities and severe dysfunction of PLM mechanosensory neurons without cell death. Proc Natl Acad Sci USA 98: 13318-13323.
  333. Pothof J, van Haaften G, Thijssen K, Kamath RS, Fraser AG, et al. (2003) Identification of genes that protect the C. elegans genome against mutations by genome-wide RNAi. Genes Dev 17: 443-448.
  334. Pulak R (2006) Techniques for analysis, sorting, and dispensing of C. elegans on the COPAS flow-sorting system. Methods Mol Biol 351: 275-286.
  335. Rosenbluth RE, Cuddeford C, Baillie DL (1985) Mutagenesis in Caenorhabditis elegans. II. A spectrum of mutational events induced with 1500 r of gamma-radiation. Genetics 109: 493-511.
  336. Rosenbluth RE, Cuddeford C, Baillie DL (1983) Mutagenesis in Caenorhabditis elegans I: A rapid eukaryotic mutagen test system using the reciprocal translocation eT1(III;V). Mutat Res 110: 39-48.
  337. Hope IA (1999) Background on Caenorhabditis elegans, C. elegans: A Practical Approach. NYOxford University Press: 1-15.
  338. Johnson TE, Hartman PS (1988) Radiation effects on life span in Caenorhabditis elegans. J Gerontol 43: B137-141.
  339. Plasterk RH (1995) Reverse genetics: from gene sequence to mutant worm. Methods Cell Biol 48: 59-80.
  340. Sambongi Y, Nagae T, Liu Y, Yoshimizu T, Takeda K, et al. (1999) Sensing of cadmium and copper ions by externally exposed ADL, ASE, and ASH neurons elicits avoidance response in Caenorhabditis elegans. Neuroreport 10: 753-757.
  341. Samii A, Nutt JG, Ransom BR (2004) Parkinson's disease. Lancet 363: 1783-1793.
  342. Schulenburg H, Muller S (2004) Natural variation in the response of Caenorhabditis elegans towards Bacillus thuringiensis. Parasitology 128: 433-443.
  343. Shatilla A, Ishchenko AA, Saparbaev M, Ramotar D (2005) Characterization of Caenorhabditis elegans exonuclease-3 and evidence that a Mg2+-dependent variant exhibits a distinct mode of action on damaged DNA. Biochemistry 44: 12835-12848.
  344. Sherwood DR, Butler JA, Kramer JM, Sternberg PW (2005) FOS-1 promotes basement-membrane removal during anchor-cell invasion in C. elegans. Cell 121: 951-962.
  345. Smialowska A, Baumeister R (2006) Presenilin function in Caenorhabditis elegans. Neurodegener Dis 3: 227-232.
  346. Sochova I, Hofman J, Holoubek I (2007) Effects of seven organic pollutants on soil nematode Caenorhabditis elegans. Environ Int 33: 798-804.
  347. Stringham-Durovic EG, Candido EPM (1994) Transgenic hsp16-lacZ strains of the soil nematode Caenorhabditis elegans as biological monitors of environmental stress. Environmental Toxicology and Chemistry 13: 1211-1220.
  348. Thiruchelvam M, Richfield EK, Baggs RB, Tank AW, Cory-Slechta DA (2000) The nigrostriatal dopaminergic system as a preferential target of repeated exposures to combined paraquat and maneb: Implications for Parkinson's disease. J Neurosci 20: 9207-9214.
  349. Tijsterman M, Pothof J, Plasterk RHA (2002) Frequent germline mutations and somatic repeat instability in DNA mismatch-repair-deficient Caenorhabditis elegans. Genetics 161: 651-660.
  350. Traunspurger W, Haitzer M, Hoss S, Beier S, Ahlf W, et al. (1997) Ecotoxicological assessment of aquatic sediments with Caenorhabditis elegans (nematoda)A method for testing liquid medium and whole-sediment samples. Environ Toxicol Chem 16: 245-250.
  351. Tseng YY, Yu CW, Liao VH (2007) Caenorhabditis elegans expresses a functional ArsA. FEBS J 274: 2566-2572.
  352. Vartiainen S, Pehkonen P, Lakso M, Nass R, Wong G (2006) Identification of gene expression changes in transgenic C. elegans overexpressing human alpha-synuclein. Neurobiol Dis 22: 477-486.
  353. Ved R, Saha S, Westlund B, Perier C, Burnam L, et al. (2005) Similar patterns of mitochondrial vulnerability and rescue induced by genetic modification of alpha-synuclein, parkin, and DJ-1 in Caenorhabditis elegans. J Biol Chem 280: 42655-42668.
  354. Wang H, Lim PJ, Yin C, Rieckher M, Vogel BE, et al. (2006) Suppression of polyglutamine-induced toxicity in cell and animal models of Huntington's disease by ubiquilin. Hum Mol Genet 15: 1025-1041.
  355. Wang YM, Pu P, Le WD (2007) ATP depletion is the major cause of MPP+ induced dopamine neuronal death and worm lethality in alpha-synuclein transgenic C. elegans. Neurosci Bull 23: 329-335.
  356. Watanabe M, Mitani N, Ishii N, Miki K (2005) A mutation in a cuticle collagen causes hypersensitivity to the endocrine disrupting chemical, bisphenol A, in Caenorhabditis elegans. Mutat Res 570: 71-80.
  357. Wei JZ, Hale K, Carta L, Platzer E, Wong C, et al. (2003) Bacillus thuringiensis crystal proteins that target nematodes. Proc Natl Acad Sci U S A 100: 2760-2765.
  358. White JG, Southgate E, Thomson JN, Brenner S (1986) The structure of the nervous system of the nematode Caenorhabditis elegans. Philos Trans R Soc Lond B Biol Sci 314: 1-340.
  359. Zhong W, Sternberg PW (2006) Genome-wide prediction of C. elegans genetic interactions. Science 311: 1481-1484.
Citation: Destefani AC (2017) Discovering Capacity in Novel Targets: C. elegans as a Model Life Form in Toxicological Research. J Clin Toxicol 7:339.

Copyright: © 2017 Destefani AC. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top