GET THE APP

Caenorhabditis elegans as a Toolkit for Studying Mammalian Aging
Journal of Nutrition & Food Sciences

Journal of Nutrition & Food Sciences
Open Access

ISSN: 2155-9600

Review Article - (2016) Volume 6, Issue 5

Caenorhabditis elegans as a Toolkit for Studying Mammalian Aging Pathways

Virendra Shukla1*#, Sangeeta Saxena1# and Sudeep Tiwari2
1Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow-226025, India, E-mail: shuklavirendra121@gmail.com
2MTN, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow-226015, India, E-mail: shuklavirendra121@gmail.com
#Contributed equally to this work
*Corresponding Author: Virendra Shukla, Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Rae Bareily Road, Lucknow-226025, India, Tel: 09415991804

Abstract

During the last few decades the free living soil nematode Caenorhabditis elegans has been highlighted as an important model organism to decipher the role of several conserved signaling pathways in longevity determination. C. elegans is a most effective in vivo model for studying aging due to its cellular complexity and high homology with mammalian biochemical and genetic pathways. Despite its apparent simplicity, lately the nematode C. elegans has been developed into an important model for biomedical research, mostly in the functional characterization of novel drug targets identified using genomics technologies. For many decades, aging was considered to be a passive, entropic process of tissue decline that occurred in a haphazard way. We know now, however, that the aging process, like so many other biological processes, is subject to regulation by classical signaling pathways viz. insulin signaling pathway, mitochondrial pathway etc. Some of these genetic pathways were first of all reported in small, short-lived organisms such as yeast, worms and flies, and a genetic alteration in same pathways turned out to extend lifespan in mammals as well. C. elegans aging mechanism provides a basis to understand how age regulation of a genetic pathway might be conserved between distantly related species. Here we review some aging pathways that are evolutionary conserved and modulates lifespan from worms to mammals viz. Insulin signaling (ILS), Dietary restriction (DR), Mitochondrial respiration and Sirtuin pathway.

Keywords: Aging; Caenorhabditis elegans; Gerontology; Insulin signaling pathway

Introduction

The nematode Caenorhabditis elegans has emerged as an important model for aging research as they are readily amenable to genetic and molecular analyses [1]. The aging processes in organisms are described by a progressive loss of physiological integrity, which leads to the weakening of biological functions and increased susceptibility towards death [2]. The primary risk factor caused by this deterioration process includes major human pathologies viz. cancer, diabetes, cardiovascular diseases and neurological disorders [3]. C. elegans has been used extensively in aging study, owing to its several advantages viz. short lifecycle (about 3 days) and short lifespan (about 3 weeks) [4]. C. elegans, the free living soil nematode was only recognized as a potential genetic model organism in the early 1960s by Sydney Brenner [5]. C. elegans strains are easy to culture and we could easily maintain on nematode growth medium (NGM) agar plates or in liquid culture seeded with E. coli OP50 bacteria (Figure 1). Furthermore, the worm’s short life cycle (approximately 3 days at 20ºC), small size (about 1-1.2 mm), high productivity, transparent appearance allow easy and quick manipulations of biogerontological studies using this as animal model as already reported by former researchers [5,6].

nutrition-food-sciences-Microscopic-view

Figure 1: Microscopic view of C. elegans.

Extensive studies on complete genome sequence, its 60-80% homology with human genes [7], knockout (KO) mutant libraries, established genetic methodologies (forward and reverse) [8], mutagenesis, carcinogenesis and recently RNA interference (RNAi) technology [9], provide a variety of options to manipulate and study C. elegans at the molecular level and excavate the associated genetic pathways, made us interested using C. elegans as a model organism to study and regulate age related genes [5,6,10,11].

Partridge reported that a single gene mutation could produce a substantial increase in its lifespan [12-14]. This led to the hypothesis that same kind of mutation and further the loss of function of the equivalent genes can extend the lifespan of mammals as most of the C. elegans genes have human orthologues [10,15], suggesting that mechanisms found to influence aging in C. elegans are likely to have a conserved role in regulating longevity in humans. Many genes that affect C. elegans life span have already been isolated and have been ordered in pathways based on genetic analysis [16]. Michael Klass conducted the first experiment that described aging in nematode C. elegans and suggested that age-related changes in this model are similar to other organisms [17]. The first gene altering aging in C.elegans was identified as age-1 by Klass and Johnson in the 1980. Mutations in this C. elegans gene increased lifespan by 50% [12,17]. The classical work on aging by Klass and Johnson (1980) transformed the field of gerontology and proved that aging is regulated at a cellular level rather than a random process. The landmark finding of age altering gene led to many more discoveries resulting in the identification of numerous genes and pathways involved in aging. Many aging pathways are identified that are evolutionary conserved and modulates lifespan from worms to mammals viz. Insulin signalling (ILS), Dietary restriction (DR), TOR signalling, Mitochondrial respiration [18], JNK signalling, Oxidative stress pathway, Sirtuin regulated signalling etc. (Figure 2) [15,19].

nutrition-food-sciences-Genetic-regulation

Figure 2: Genetic regulation of lifespan in C. elegans by evolutionary conserved mechanism.

The main pathway that regulates life span is reported to be insulinlike signalling (ILS) pathway which effects longevity in worms [15]. Mutations in various genes of ILS pathway have resulted influencing lifespan in flies, mice and also in human longevity.

Insulin Signalling Pathway (ILS)

Insulin signalling pathway is an evolutionarily conserved pathway, modulating organism’s lifespan and regulating several cellular processes such as stress resistance against abiotic factors, immunity against biotic factors and aging [20,21]. The impact of this pathway was first identified using the invertebrate model C. elegans [13,17,22]. Insulin like growth factor (IGF)/ILS pathways have been shown to influence lifespan in vertebrates as well, including human beings (Figure 3) [20,23,24]. In C. elegans , the IIS signalling pathway consists of various proteins that are encoded by the genes daf-2, age-1, akt-1, akt-2, daf-16 and daf-18 . The gene daf-2 encodes a homolog of the mammalian insulin/IGF-I receptor (Figure 3) [25] while age-1 encodes the catalytic p110 subunit of phosphoinositide-3-OH kinase (PI3K) situated downstream of DAF-2 [26]. Mutations in these genes (age-1 and daf-2 ) increase the longevity and stress resistance of nematodes in daf-16 dependent manner. In this pathway, stimulation of daf-2 receptor activates the phosphatidylinositol 3-kinase (PI 3-kinase). AGE 1 that consists of a p55-like regulatory subunit [27] and a p110 catalytic subunit [28] that is antagonized by DAF-18. Thereafter, PIP3 signal activates the AKT/PKB kinase homologue PDK-1, which phosphorylates AKT-1, AKT-2 and SGK-1. The AKT proteins are responsible for dauer formation as well as for lifespan extension while SGK-1 proteins regulate development, stress response and longevity in worms. These kinases then phosphorylate the transcription factor DAF-16 and inactivate it by cytoplasmic retention [6,27,29-34].

nutrition-food-sciences-Schematic-diagram

Figure 3: Schematic diagram of the ILS/IGF pathway in C. elegans (A) Inactivated and (B) Activated pathway.

The sub-cellular DAF-16 cytoplasmic localization is regulated by the formation of a protein complex between 14-3-3 FTT-2 and DAF-16, a mechanism that appears to be conserved in mammals [35]. Mutations in daf-2 or other upstream components of insulin signalling pathway such as age-1, akt-1 that result in a reduction in IGF-I signalling cause dephosphorylation of DAF-16 and its subsequent translocation into the nucleus. Nucleus localized DAF-16 regulates a number of target genes that have been identified to be involved in various mechanism such as dauer formation, metabolism, development, stress response, detoxification and other signalling (Figure 3B) [36-42].

Apart from insulin signalling pathway and its molecular players, many genes and genetic signalling pathways have been discovered in worms which are related to lifespan extension. A few important genes/ pathways discovered are: Dietary restriction (DR), SIR2 deacetylase activity (probably with importance in DR), JNK-signaling, TOR, mitochondrial mechanisms, oxidative stress signaling and others [43].

Dietary Restriction (DR) Pathway

Dietary restriction is the reduction of dietary intake without starvation that substantially increases lifespan [44]. This term is also known as caloric restriction (CR). During different time interval similar effects have been observed from lower to higher organisms such as baker’s yeast (Saccharomyces cerevisiae), fruitfly (Drosophila melanogaster ), mice (Mus musculus ), dogs (Canis lupus familiaris) and rhesus monkeys (Macaca mulatta ) [45,46]. Restricting food intake of rodents can extend their lifespan, and this extension is specifically due to a reduction of caloric intake. Reducing calories can also extend life span in other organisms, such as yeast and C. elegans [47,48]. The first study report DR mediated longevity in C. elegans involved dilution of food bacteria (E. coli OP50) in buffer [49,50]. The decreasing concentration of E. coli caused lifespan extension and reduction in worm’s fertility. Another approach showed same result where reducing the nutrient content in the agar plate showed an inverse relationship between lifespan and bacterial quantity [36]. The DR studies in C. elegans have been to correlate the biology of DR to the identified various aging genes/pathways viz. eat-2, sir2.1, pha-4 etc. [51,52].

Sirtuin Signaling

For the study of aging and longevity SIRT1 is most extensively studied and implicated as a key mediator in Dietary restriction [53]. Sirtuins are highly conserved NAD+-dependent protein deacetylases that identified early in yeast known as silent information regulator (Sir) [54]. A number of studies suggest that over expression of Sir2 increase lifespan in many models organism such as yeast, C. elegans and Drosophila [19]. In mammals seven SIRT proteins (SIRT1-7) are reported that are either NAD-dependent deacetylases or protein ADPribosyltransferases and display diversity of functions [55]. In C. elegans, over expression of the sirtuin gene sir-2.1 causes longevity by activating DAF-16/FOXO [56,57]. SIR-2.1 is probably activating transcription factor DAF-16 by deacetylation. As it is reported that in response to oxidative stress mammalian SIRT1 is known to deacetylate FOXO proteins therefore it likely shifts their target towards the antioxidant genes which are play a major role in stress resistance [56]. In the C. elegans oxidative stress conditions may be stimulates the binding of SIR-2.1 towards DAF-16 [57] and cause lifespan extension in a sir-2 dependent and daf-16- dependent manner [58]. The findings advocate that sirtuins can deacetylate FOXO proteins directly, as well as it has been reported that insulin/IGF-1 pathway mutants do not require sir-2.1 for longevity. This phenomenon reveals that sirtuins may influence DAF-16/FOXO transcription factor and also enhance lifespan independently of insulin/IGF-1 signalling in worms. The mechanism of DR have been determined through developmental delays, reduced metabolic rate, attenuated glucocorticoid pathways [59], diminished fat levels, decreased ROS levels, increased rate of cell survival and changes in protein turnover rates.

Mitochondria and Metabolic Control of Aging Pathway

The free radical theory of aging is the most widely accepted theory that explored casual link between the rate of aging and free radical damage [60]. As the mitochondrion is known for ATP generation and one of the primary sites of ROS production in the cell, therefore these organelles are very important and are assumed to have major impact on the aging process [61,62]. Mutations in several genes that affect mitochondrial function probably have an impact on ROS generation (either increasing or decreasing ROS). The point mutations in numerous genes are directly associated with ETC function (clk-1, isp-1, and nuo-6 ) that can extend lifespan [18]. Mutations in any one of these genes results in the extended lifespan, cell cycle length, and a slowing down of development and behavioural activity (pharyngeal pumping, defecating, egg laying, and moving). The genetic mutation where both deletions and point mutations have been found to increase lifespan was first reported in the clk-1 [63]. The clk-1 gene also affects aging and several other physiological rates in C. elegans [64] and encodes an enzyme that is responsible for biosynthesis of ubiqunone [63,65,66] an essential cofactor in numerous redox reactions, including mitochondrial respiration, as a membrane antioxidant and an oxygen sensor [67]. The mitochondrial protein CLK-1 is conserved among eukaryotes and recognized as a hydroxylase involved in the ubiquinone biosynthesis (UQ9) in C. elegans [68-70]. In clk-1 mutants reduced respiration suggests that longevity is directly linked with decrease mitochondrial function which is observed in many species.

Conclusion

In conclusion, this review found that studies in the model organism C. elegans have identified genes/pathways that may regulate lifespan in a conserved manner. Recent work has firmly established the role of C. elegans in the study of aging process. There is now strong evidence for the close evolutionary conservation of lifespan regulating mechanism in a number of model organisms, supporting a similar control in human also. Comparison of aging transcriptional profiles in worms, flies, mice and humans provides a quantitative, global view of the overall relatedness of the aging process across different species. These results provide a view of the relative proportion of the aging process that is specific to humans rather than shared across animals. The frontier of aging research now lies in the light of this information to mammals and in particular to uncover the processes most important in human aging. It is likely that the framework that has emerged from the study of model organism C. elegans and the pathways involved will provide a basis for an understanding of aging which can be further applied on human as well.

Acknowledgment

Authors are grateful to Babasaheb Bhimrao Ambedkar University (A Central University) Lucknow, India for providing the necessary infrastructure. One of the author (VS) is grateful to Dr. D.S. Kothari post doctoral fellowship from University Grant Commission, New Delhi (BL/1415/0241).

References

  1. Tissenbaum HA (2015) Using C. elegans for aging research. InvertebrReprodDev 59: 59-63.
  2. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153: 1194-1217.
  3. Campisi G, Chiappelli M, De Martinis M, Franco V, Ginaldi L, et al. (2009) Pathophysiology of age-related diseases. Immun Ageing 6: 12.
  4. Guarente L, Kenyon C (2000) Genetic pathways that regulate ageing in model organisms. Nature 408: 255-262.
  5. Brenner S (1974) The genetics of Caenorhabditiselegans. Genetics 77: 71-94.
  6. Braeckman BP, Vanfleteren JR (2007) Genetic control of longevity in C. elegans. ExpGerontol 42: 90-98.
  7. Baumeister R, Ge L (2002) The worm in us-Caenorhabditiselegans as a model of human disease. Trends Biotechnol 20: 147-148.
  8. Coulson A, Sulston J, Brenner S, Karn J (1986) Toward a physical map of the genome of the nematode Caenorhabditiselegans. ProcNatlAcadSci U S A 83: 7821-7825.
  9. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, et al. (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditiselegans. Nature 391: 806-811.
  10. Kenyon C (2011) The first long-lived mutants: discovery of the insulin/IGF-1 pathway for ageing. Philos Trans R SocLond B BiolSci 366: 9-16.
  11. Rodriguez M, Snoek LB, De Bono M, Kammenga JE (2013) Worms under stress: C. elegans stress response and its relevance to complex human disease and aging. Trends Genet 29: 367-374.
  12. Friedman DB, Johnson TE (1988) A mutation in the age-1 gene in Caenorhabditiselegans lengthens life and reduces hermaphrodite fertility. Genetics 118: 75-86.
  13. Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R (1993) A C. elegans mutant that lives twice as long as wild type. Nature 366: 461-464.
  14. Partridge L (2010) The new biology of ageing. Philos Trans R SocLond B BiolSci 365: 147-154.
  15. Friedman DB, Johnson TE (1988) Three mutants that extend both mean and maximum life span of the nematode, Caenorhabditiselegans, define the age-1 gene. J Gerontol 43: B102-B109.
  16. Klass MR (1983) A method for the isolation of longevity mutants in the nematode Caenorhabditiselegans and initial results. Mech Ageing Dev 22: 279-286.
  17. Ishii N, Fujii M, Hartman PS, Tsuda M, Yasuda K, et al. (1998) A mutation in succinate dehydrogenase cytochrome b causes oxidative stress and ageing in nematodes. Nature 394:694-697.
  18. Tissenbaum HA, Guarente L (2001) Increased dosage of a sir-2 gene extends lifespan in Caenorhabditiselegans. Nature 410: 227-230.
  19. Holzenberger M, Dupont J, Ducos B, Leneuve P, Géloën A, et al. (2003) IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421: 182-187.
  20. Rincon M, Rudin E, Barzilai N (2005) The insulin/IGF-1 signaling in mammals and its relevance to human longevity. ExpGerontol 40: 873-877.
  21. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G (1997) daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditiselegans. Science 277: 942-946.
  22. Brown-Borg HM, Borg KE, Meliska CJ, Bartke A (1996) Dwarf mice and the ageing process. Nature 384: 33.
  23. Blüher M1, Kahn BB, Kahn CR (2003) Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 299: 572-574.
  24. Brown WT (1979) Human mutations affecting aging-a review. Mech Ageing Dev 9: 325-336.
  25. Garigan D, Hsu AL, Fraser AG, Kamath RS, Ahringer J, et al. (2002) Genetic analysis of tissue aging in Caenorhabditiselegans: a role for heat-shock factor and bacterial proliferation. Genetics 161:1101-1112.
  26. Iser WB, Gami MS, Wolkow CA (2007) Insulin signaling in Caenorhabditiselegans regulates both endocrine-like and cell-autonomous outputs. DevBiol 303: 434-447.
  27. Morris JZ, Tissenbaum HA, Ruvkun G (1996) A phosphatidylinositol-3-OH kinase family member regulating longevity and diapause in Caenorhabditiselegans. Nature 382: 536-539.
  28. Ogg S, Ruvkun G (1998) The C. elegans PTEN homolog, DAF-18, acts in the insulin receptor-like metabolic signaling pathway. Mol Cell 2: 887-893.
  29. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, et al. (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857-868.
  30. Finch CE, Ruvkun G (2001) The genetics of aging. Annu Rev Genomics Hum Genet 2: 435-462.
  31. Lin K, Hsin H, Libina N, Kenyon C (2001) Regulation of the Caenorhabditiselegans longevity protein DAF-16 by insulin/IGF-1 and germlinesignaling. Nat Genet 28: 139-145.
  32. Chávez V, Mohri-Shiomi A, Maadani A, Vega LA, Garsin DA (2007) Oxidative stress enzymes are required for DAF-16-mediated immunity due to generation of reactive oxygen species by Caenorhabditiselegans. Genetics 176: 1567-1577.
  33. Li J, Tewari M, Vidal M, Lee SS (2007) The 14-3-3 protein FTT-2 regulates DAF-16 in Caenorhabditiselegans. DevBiol 301: 82-91.
  34. Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, et al. (2003) Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditiselegans. Nature 424: 277-283.
  35. Lee SS, Kennedy S, Tolonen AC, Ruvkun G (2003) DAF-16 target genes that control C. elegans life-span and metabolism. Science 300: 644-647.
  36. Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, et al. (2006) Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet 38: 251-257.
  37. Dong MQ, Venable JD, Au N, Xu T, Park SK, et al. (2007) Quantitative mass spectrometry identifies insulin signaling targets in C. elegans. Science 317: 660-663.
  38. McElwee JJ, Schuster E, Blanc E, Piper MD, Thomas JH, et al. (2007) Evolutionary conservation of regulated longevity assurance mechanisms. Genome Biol 8: R132.
  39. Shukla V, Yadav D, Phulara SC, Gupta MM, Saikia SK, et al. (2012) Longevity-promoting effects of 4-hydroxy-E-globularinin in Caenorhabditiselegans. Free RadicBiol Med 53: 1848-1856.
  40. Shukla V, Phulara SC, Yadav D, Tiwari S, Kaur S, et al. (2012) Iridoid compound 10-O-trans-p-coumaroylcatalpol extends longevity and reduces a synuclein aggregation in Caenorhabditiselegans. CNS NeurolDisord Drug Targets 11:984-992.
  41. Schaffitzel E, Hertweck M (2006) Recent aging research in Caenorhabditiselegans. ExpGerontol 41: 557-563.
  42. McCay CM, Crowell MF, Maynard LA (1989) The effect of retarded growth upon the length of life span and upon the ultimate body size. Nutrition 5: 155-171.
  43. Walker DS (2005) C. eleganscalexcitin 1 functions in associative learning and serotonin mediated responses. International Worm Meeting, pp: 246.
  44. Lin SJ, Kaeberlein M, Andalis AA, Sturtz LA, Defossez PA, et al. (2002) Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. Nature 418: 344-348.
  45. GreerEL, Brunet A (2009) Different dietary restriction regimens extend lifespan by both independent and overlapping genetic pathways in C. elegans. Aging Cell 8: 113-127.
  46. Lakowski B, Hekimi S (1998) The genetics of caloric restriction in Caenorhabditiselegans. ProcNatlAcadSci USA 95: 13091-13096.
  47. Klass MR (1977) Aging in the nematode Caenorhabditiselegans: major biological and environmental factors influencing life span. Mech Ageing Dev 6: 413-429.
  48. Hosono R, Nishimoto S, Kuno S (1989) Alterations of life span in the nematode Caenorhabditiselegans under monoxenic culture conditions. ExpGerontol 24: 251-264.
  49. Avery L (1993) The genetics of feeding in Caenorhabditiselegans. Genetics 133: 897-917.
  50. McKay JP, Raizen DM, Gottschalk A, Schafer WR, Avery L (2004) Eat-2 and eat-18 are required for nicotinic neurotransmission in the Caenorhabditiselegans pharynx. Genetics 166: 161-169.
  51. Haigis MC, Sinclair DA (2010) Mammalian sirtuins: biological insights and disease relevance. Annu Rev Pathol 5: 253-295.
  52. Viswanathan M, Tissenbaum HA (2013) C. eleganssirtuins. Methods Mol Biol1077:39-56
  53. Kalaany NY, Sabatini DM (2009) Tumours with PI3K activation are resistant to dietary restriction. Nature 458: 725-731.
  54. Kenyon C (2005) The plasticity of aging: insights from long-lived mutants. Cell 120: 449-460.
  55. Berdichevsky A, Viswanathan M, Horvitz HR, Guarente L (2006) C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span. Cell 125: 1165-1177.
  56. Heidler T, Hartwig K, Daniel H, Wenzel U (2010) Caenorhabditiselegans lifespan extension caused by treatment with an orally active ROS-generator is dependent on DAF-16 and SIR-2.1. Biogerontology 11: 183-195.
  57. Masoro EJ, Austad SN (1996) The evolution of the antiaging action of dietary restriction: a hypothesis. J Gerontol A BiolSci Med Sci 51: B387-B391.
  58. Harman D (1956) Aging: a theory based on free radical and radiation chemistry. J Gerontol 11:298-300.
  59. Murphy MP (2009) How mitochondria produce reactive oxygen species. Biochem J 417: 1-13.
  60. Sebastián D, Acín-Pérez R, Morino K (2016) Mitochondrial Health in Aging and Age-Related Metabolic Disease. Oxidative Medicine and Cellular Longevity 2016: 5831538.
  61. Ewbank JJ, Barnes TM, Lakowski B, Lussier M, Bussey H, et al. (1997) Structural and functional conservation of the Caenorhabditiselegans timing gene clk-1. Science 275: 980-983.
  62. Wong A, Boutis P, Hekimi S (1995) Mutations in the clk-1 gene of Caenorhabditiselegans affect developmental and behavioral timing. Genetics 139: 1247-1259.
  63. Marbois BN, Clarke CF (1996) The COQ7 gene encodes a protein in saccharomyces cerevisiae necessary for ubiquinone biosynthesis. J BiolChem 271: 2995-3004.
  64. Miyadera H, Amino H, Hiraishi A, Taka H, Murayama K, et al. (2001) Altered quinone biosynthesis in the long-lived clk-1 mutants of Caenorhabditiselegans. J BiolChem 276: 7713-7716.
  65. Georgellis D, Kwon O, Lin EC (2001) Quinones as the redox signal for the arc two-component system of bacteria. Science 292: 2314-2316.
  66. Branicky R, Shibata Y, Feng J, Hekimi S (2001) Phenotypic and suppressor analysis of defecation in clk-1 mutants reveals that reaction to changes in temperature is an active process in Caenorhabditiselegans. Genetics 159: 997-1006.
  67. Hekimi S, Bénard C, Branicky R, Burgess J, Hihi AK, et al. (2001) Why only time will tell. Mech Ageing Dev 122: 571-594.
  68. Jonassen T, Larsen PL, Clarke CF (2001) A dietary source of coenzyme Q is essential for growth of long-lived Caenorhabditiselegansclk-1 mutants. ProcNatlAcadSci USA 98: 421-426.
Citation: Shukla V, Saxena S, Tiwari S (2016) Caenorhabditis Elegans as a Toolkit for Studying Mammalian Aging Pathways. J Nutr Food Sci 6:542.

Copyright: © 2016 Shukla V, et al. 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