201
|
Lemieux GA, Ashrafi K. Insights and challenges in using C. elegans for investigation of fat metabolism. Crit Rev Biochem Mol Biol 2014; 50:69-84. [PMID: 25228063 DOI: 10.3109/10409238.2014.959890] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
C. elegans provides a genetically tractable system for deciphering the homeostatic mechanisms that underlie fat regulation in intact organisms. Here, we provide an overview of the recent advances in the C. elegans fat field with particular attention to studies of C. elegans lipid droplets, the complex links between lipases, autophagy, and lifespan, and analyses of key transcriptional regulatory mechanisms that coordinate lipid homeostasis. These studies demonstrate the ancient origins of mammalian and C. elegans fat regulatory pathways and highlight how C. elegans is being used to identify and analyze novel lipid pathways that are then shown to function similarly in mammals. Despite its many advantages, study of fat regulation in C. elegans is currently faced with a number of conceptual and methodological challenges. We critically evaluate some of the assumptions in the field and highlight issues that we believe should be taken into consideration when interpreting lipid content data in C. elegans.
Collapse
Affiliation(s)
- George A Lemieux
- Department of Physiology, University of California , San Francisco, CA , USA
| | | |
Collapse
|
202
|
Zhou QJ, Yang Y, Guo XL, Duan LJ, Chen XQ, Yan BL, Zhang HL, Du AF. Expression of Caenorhabditis elegans-expressed Trans-HPS, partial aminopeptidase H11 from Haemonchus contortus. Exp Parasitol 2014; 145:87-98. [PMID: 25128369 DOI: 10.1016/j.exppara.2014.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/29/2014] [Accepted: 08/04/2014] [Indexed: 01/26/2023]
Abstract
Aminopeptidase H11 present in the surface of intestine microvilli in Haemonchus contortus was identified as the most effective antigen candidate. However, its recombinant forms produced in Escherichiacoli, insect cells and yeast could not provide promising protection against H. contortus challenge, probably due to the inappropriate glycosylation and/or conformational folding. Herein, partial H11 containing the potential zinc-binding domain and two predicted glycosylation sites (nt 1 bp-1710 bp, Trans-HPS) was subcloned downstream of 5' flanking region of Caenorhabditis elegans cpr-1 gene in pPD95.77 vector, with the deletion of GFP gene. The recombinant was expressed in C. elegans and verified by blotting with anti-H11 and anti-Trans-HPS rabbit polyclonal antibodies and anti-His monoclonal antibody. Stably inherited Trans-HPS in worm descendants was achieved by integration using UV irradiation. Immunization with the crude Trans-HPS extracted from transgenic worms resulted in 37.71% reduction in faecal egg counts (FEC) (P<0.05) and 24.91% reduction in worm burden, but an upward curve with moderate rate of daily FEC in goats. These results suggested an apparent delay against H. contortus egg-laying in goats, which differed from that with bacteria-origin form of partial H11 (nt 670 bp-1710 bp, HPS) (26.04% reduction in FEC and 18.46% reduction in worm burden). These findings indicate the feasibility of sufficient C. elegans-expressed H11 for the immunological research and vaccine development.
Collapse
Affiliation(s)
- Qian-Jin Zhou
- School of Marine Science, Ningbo University, Ningbo 315211, China; Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yi Yang
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Lu Guo
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Li-Jun Duan
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue-Qiu Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Bao-Long Yan
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hong-Li Zhang
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ai-Fang Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
203
|
Dwyer DS, Aamodt E, Cohen B, Buttner EA. Drug elucidation: invertebrate genetics sheds new light on the molecular targets of CNS drugs. Front Pharmacol 2014; 5:177. [PMID: 25120487 PMCID: PMC4112795 DOI: 10.3389/fphar.2014.00177] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/09/2014] [Indexed: 02/02/2023] Open
Abstract
Many important drugs approved to treat common human diseases were discovered by serendipity, without a firm understanding of their modes of action. As a result, the side effects and interactions of these medications are often unpredictable, and there is limited guidance for improving the design of next-generation drugs. Here, we review the innovative use of simple model organisms, especially Caenorhabditis elegans, to gain fresh insights into the complex biological effects of approved CNS medications. Whereas drug discovery involves the identification of new drug targets and lead compounds/biologics, and drug development spans preclinical testing to FDA approval, drug elucidation refers to the process of understanding the mechanisms of action of marketed drugs by studying their novel effects in model organisms. Drug elucidation studies have revealed new pathways affected by antipsychotic drugs, e.g., the insulin signaling pathway, a trace amine receptor and a nicotinic acetylcholine receptor. Similarly, novel targets of antidepressant drugs and lithium have been identified in C. elegans, including lipid-binding/transport proteins and the SGK-1 signaling pathway, respectively. Elucidation of the mode of action of anesthetic agents has shown that anesthesia can involve mitochondrial targets, leak currents, and gap junctions. The general approach reviewed in this article has advanced our knowledge about important drugs for CNS disorders and can guide future drug discovery efforts.
Collapse
Affiliation(s)
- Donard S. Dwyer
- Department of Psychiatry–Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-ShreveportShreveport, LA, USA
| | - Eric Aamodt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-ShreveportShreveport, LA, USA
| | - Bruce Cohen
- Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
- Mailman Research Center, McLean HospitalBelmont, MA, USA
| | - Edgar A. Buttner
- Mailman Research Center, McLean HospitalBelmont, MA, USA
- Department of Neurology–Department of Psychiatry, McLean Hospital, Harvard Medical SchoolBelmont, MA, USA
| |
Collapse
|
204
|
Kinnunen TK. Combinatorial roles of heparan sulfate proteoglycans and heparan sulfates in Caenorhabditis elegans neural development. PLoS One 2014; 9:e102919. [PMID: 25054285 PMCID: PMC4108370 DOI: 10.1371/journal.pone.0102919] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 06/25/2014] [Indexed: 02/06/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) play critical roles in the development and adult physiology of all metazoan organisms. Most of the known molecular interactions of HSPGs are attributed to the structurally highly complex heparan sulfate (HS) glycans. However, whether a specific HSPG (such as syndecan) contains HS modifications that differ from another HSPG (such as glypican) has remained largely unresolved. Here, a neural model in C. elegans is used to demonstrate for the first time the relationship between specific HSPGs and HS modifications in a defined biological process in vivo. HSPGs are critical for the migration of hermaphrodite specific neurons (HSNs) as genetic elimination of multiple HSPGs leads to 80% defect of HSN migration. The effects of genetic elimination of HSPGs are additive, suggesting that multiple HSPGs, present in the migrating neuron and in the matrix, act in parallel to support neuron migration. Genetic analyses suggest that syndecan/sdn-1 and HS 6-O-sulfotransferase, hst-6, function in a linear signaling pathway and glypican/lon-2 and HS 2-O-sulfotransferase, hst-2, function together in a pathway that is parallel to sdn-1 and hst-6. These results suggest core protein specific HS modifications that are critical for HSN migration. In C. elegans, the core protein specificity of distinct HS modifications may be in part regulated at the level of tissue specific expression of genes encoding for HSPGs and HS modifying enzymes. Genetic analysis reveals that there is a delicate balance of HS modifications and eliminating one HS modifying enzyme in a compromised genetic background leads to significant changes in the overall phenotype. These findings are of importance with the view of HS as a critical regulator of cell signaling in normal development and disease.
Collapse
Affiliation(s)
- Tarja K. Kinnunen
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom, and Department of Biology, University of Huddersfield, Huddersfield, United Kingdom
- * E-mail:
| |
Collapse
|
205
|
Pilon M. Developmental genetics of the Caenorhabditis elegans pharynx. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2014; 3:263-80. [PMID: 25262818 PMCID: PMC4314705 DOI: 10.1002/wdev.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/03/2014] [Accepted: 04/15/2014] [Indexed: 11/07/2022]
Abstract
The Caenorhabditis elegans pharynx is a rhythmically pumping organ composed initially of 80 cells that, through fusions, amount to 62 cells in the adult worm. During the first 100 min of development, most future pharyngeal cells are born and gather into a double-plate primordium surrounded by a basal lamina. All pharyngeal cells express the transcription factor PHA-4, of which the concentration increases throughout development, triggering a sequential activation of genes with promoters responding differentially to PHA-4 protein levels. The oblong-shaped pharyngeal primordium becomes polarized, many cells taking on wedge shapes with their narrow ends toward the center, hence forming an epithelial cyst. The primordium then elongates, and reorientations of the cells at the anterior and posterior ends form the mouth and pharyngeal-intestinal openings, respectively. The 20 pharyngeal neurons establish complex but reproducible trajectories using 'fishing line' and growth cone-driven mechanisms, and the gland cells also similarly develop their processes. The genetics behind many fate decisions and morphogenetic processes are being elucidated, and reveal the pharynx to be a fruitful model for developmental biologists.
Collapse
Affiliation(s)
- Marc Pilon
- Department of Chemistry and Molecular Biology, University of GothenburgGothenburg, Sweden
| |
Collapse
|
206
|
Cunningham KA, Bouagnon AD, Barros AG, Lin L, Malard L, Romano-Silva MA, Ashrafi K. Loss of a neural AMP-activated kinase mimics the effects of elevated serotonin on fat, movement, and hormonal secretions. PLoS Genet 2014; 10:e1004394. [PMID: 24921650 PMCID: PMC4055570 DOI: 10.1371/journal.pgen.1004394] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 04/07/2014] [Indexed: 12/30/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved master regulator of metabolism and a therapeutic target in type 2 diabetes. As an energy sensor, AMPK activity is responsive to both metabolic inputs, for instance the ratio of AMP to ATP, and numerous hormonal cues. As in mammals, each of two genes, aak-1 and aak-2, encode for the catalytic subunit of AMPK in C. elegans. Here we show that in C. elegans loss of aak-2 mimics the effects of elevated serotonin signaling on fat reduction, slowed movement, and promoting exit from dauer arrest. Reconstitution of aak-2 in only the nervous system restored wild type fat levels and movement rate to aak-2 mutants and reconstitution in only the ASI neurons was sufficient to significantly restore dauer maintenance to the mutant animals. As in elevated serotonin signaling, inactivation of AAK-2 in the ASI neurons caused enhanced secretion of dense core vesicles from these neurons. The ASI neurons are the site of production of the DAF-7 TGF-β ligand and the DAF-28 insulin, both of which are secreted by dense core vesicles and play critical roles in whether animals stay in dauer or undergo reproductive development. These findings show that elevated levels of serotonin promote enhanced secretions of systemic regulators of pro-growth and differentiation pathways through inactivation of AAK-2. As such, AMPK is not only a recipient of hormonal signals but can also be an upstream regulator. Our data suggest that some of the physiological phenotypes previously attributed to peripheral AAK-2 activity on metabolic targets may instead be due to the role of this kinase in neural serotonin signaling.
Collapse
Affiliation(s)
- Katherine A. Cunningham
- Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Aude D. Bouagnon
- Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Alexandre G. Barros
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lin Lin
- Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Leandro Malard
- Departamento de Física, Instituto de Ciências Exatas da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marco Aurélio Romano-Silva
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
207
|
Abstract
Nearly all animals are capable of sensing changes in environmental oxygen (O2) and carbon dioxide (CO2) levels, which can signal the presence of food, pathogens, conspecifics, predators, or hosts. The free-living nematode Caenorhabditis elegans is a powerful model system for the study of gas sensing. C. elegans detects changes in O2 and CO2 levels and integrates information about ambient gas levels with other internal and external cues to generate context-appropriate behavioral responses. Due to its small nervous system and amenability to genetic and genomic analyses, the functional properties of its gas-sensing microcircuits can be dissected with single-cell resolution, and signaling molecules and natural genetic variations that modulate gas responses can be identified. Here, we discuss the neural basis of gas sensing in C. elegans, and highlight changes in gas-evoked behaviors in the context of other sensory cues and natural genetic variations. We also discuss gas sensing in other free-living nematodes and parasitic nematodes, focusing on how gas-sensing behavior has evolved to mediate species-specific behavioral requirements.
Collapse
|
208
|
Longevity manipulations differentially affect serotonin/dopamine level and behavioral deterioration in aging Caenorhabditis elegans. J Neurosci 2014; 34:3947-58. [PMID: 24623772 DOI: 10.1523/jneurosci.4013-13.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aging is accompanied with behavioral and cognitive decline. Changes in the neurotransmitter level are associated with the age-related behavioral deterioration, but whether well-known longevity manipulations affect the function of neurotransmitter system in aging animals is largely unclear. Here we report that serotonin (5-HT) and dopamine (DA) level decrease with age in C. elegans. The reduction results in downregulation of the activity of neurons controlled by 5-HT/DA signaling, and deterioration of some important behaviors, including pharyngeal pumping, food-induced slowing responses, and male mating. Longevity manipulations differentially affect the age-related decline in neuronal level of 5-HT/DA. The reduction and resultant behavioral deterioration occur in long-lived worms with defective insulin signaling [daf-2(e1370), age-1(hx546)] or mitochondria function [isp-1(qm150), tpk-1(qm162)], but not in long-lived worms with dietary restriction eat-2(ad1116). A reduced expression level of dopa decarboxylase BAS-1, the shared enzyme for 5-HT/DA synthesis, is responsible for the decline in 5-HT/DA levels. RNAi assay revealed that the sustained 5-HT/DA level in neurons of aged eat-2(ad1116) worms requires PHA-4 and its effectors superoxide dismutases and catalases, suggesting the involvement of reactive oxygen species in the 5-HT/DA decline. Furthermore, we found that elevating 5-HT/DA ameliorates age-related deterioration of pharyngeal pumping, food-induced slowing responses, and male mating in both wild-type and daf-2(e1370) worms. Together, dietary restriction preserves healthy behaviors in aged worms at least partially by sustaining a high 5-HT/DA level, and elevating the 5-HT/DA level in wild-type and daf-2(e1370) worms improves their behaviors during aging.
Collapse
|
209
|
Glebov K, Voronezhskaya EE, Khabarova MY, Ivashkin E, Nezlin LP, Ponimaskin EG. Mechanisms underlying dual effects of serotonin during development of Helisoma trivolvis (Mollusca). BMC DEVELOPMENTAL BIOLOGY 2014; 14:14. [PMID: 24625099 PMCID: PMC4007640 DOI: 10.1186/1471-213x-14-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 02/21/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Serotonin (5-HT) is well known as widely distributed modulator of developmental processes in both vertebrates and invertebrates. It is also the earliest neurotransmitter to appear during neuronal development. In aquatic invertebrates, which have larvae in their life cycle, 5-HT is involved in regulation of stages transition including larval metamorphosis and settlement. However, molecular and cellular mechanisms underlying developmental transition in aquatic invertebrate species are yet poorly understood. Earlier we demonstrated that in larvae of freshwater molluscs and marine polychaetes, endogenous 5-HT released from the neurons of the apical sensory organ (ASO) in response to external stimuli retarded larval development at premetamorphic stages, and accelerated it at metamorphic stages. Here we used a freshwater snail Helisoma trivolvis to study molecular mechanisms underlying these dual developmental effects of 5-HT. RESULTS Larval development of H. trivolvis includes transition from premetamorphic to metamorphic stages and shares the main features of metamorphosis with free-swimming aquatic larvae. Three types of 5-HT receptors (5-HT1-, 5-HT4- and 5-HT7-like) are functionally active at premetamorphic (trochophore, veliger) and metamorphic (veliconcha) stages, and expression patterns of these receptors and respective G proteins undergo coordinated changes during development. Stimulation of these receptors modulated cAMP-dependent regulation of cell divisions. Expression of 5-HT4- and 5-HT7-like receptors and their downstream Gs protein was down-regulated during the transition of pre- to metamorphic stage, while expression of 5-HT1 -like receptor and its downstream Gi protein was upregulated. In accordance with relative amount of these receptors, stimulation of 5-HTRs at premetamorphic stages induces developmental retardation, while their stimulation at metamorphic stages induces developmental acceleration. CONCLUSIONS We present a novel molecular mechanism that underlies stage-specific changes in developmental tempo of H. trivolvis larvae in response to endogenous 5-HT produced by the neurons of the ASO. We suggest that consecutive changes in expression patterns of different receptors and their downstream partners in the course of larval development represent the molecular base of larval transition from premetamorphic (non-competent) to metamorphic (competent) state.
Collapse
Affiliation(s)
| | | | | | | | | | - Evgeni G Ponimaskin
- DFG-Research Center Molecular Physiology of the Brain (CMPB), Göttingen, Germany.
| |
Collapse
|
210
|
Mowrey WR, Bennett JR, Portman DS. Distributed effects of biological sex define sex-typical motor behavior in Caenorhabditis elegans. J Neurosci 2014; 34:1579-91. [PMID: 24478342 PMCID: PMC3905135 DOI: 10.1523/jneurosci.4352-13.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 12/24/2022] Open
Abstract
Sex differences in shared behaviors (for example, locomotion and feeding) are a nearly universal feature of animal biology. Though these behaviors may share underlying neural programs, their kinematics can exhibit robust differences between males and females. The neural underpinnings of these differences are poorly understood because of the often-untested assumption that they are determined by sex-specific body morphology. Here, we address this issue in the nematode Caenorhabditis elegans, which features two sexes with distinct body morphologies but similar locomotor circuitry and body muscle. Quantitative behavioral analysis shows that C. elegans and related nematodes exhibit significant sex differences in the dynamics and geometry of locomotor body waves, such that the male is generally faster. Using a recently proposed model of locomotor wave propagation, we show that sex differences in both body mechanics and the intrinsic dynamics of the motor system can contribute to kinematic differences in distinct mechanical contexts. By genetically sex-reversing the properties of specific tissues and cells, however, we find that sex-specific locomotor frequency in C. elegans is determined primarily by the functional modification of shared sensory neurons. Further, we find that sexual modification of body wall muscle together with the nervous system is required to alter body wave speed. Thus, rather than relying on a single focus of modification, sex differences in motor dynamics require independent modifications to multiple tissue types. Our results suggest shared motor behaviors may be sex-specifically optimized though distributed modifications to several aspects of morphology and physiology.
Collapse
Affiliation(s)
| | | | - Douglas S. Portman
- Center for Neural Development and Disease
- Department of Biomedical Genetics, and
- Department of Biology, University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
211
|
Barros AGDA, Bridi JC, de Souza BR, de Castro Júnior C, de Lima Torres KC, Malard L, Jorio A, de Miranda DM, Ashrafi K, Romano-Silva MA. Dopamine signaling regulates fat content through β-oxidation in Caenorhabditis elegans. PLoS One 2014; 9:e85874. [PMID: 24465759 PMCID: PMC3899111 DOI: 10.1371/journal.pone.0085874] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 12/06/2013] [Indexed: 11/26/2022] Open
Abstract
The regulation of energy balance involves an intricate interplay between neural mechanisms that respond to internal and external cues of energy demand and food availability. Compelling data have implicated the neurotransmitter dopamine as an important part of body weight regulation. However, the precise mechanisms through which dopamine regulates energy homeostasis remain poorly understood. Here, we investigate mechanisms through which dopamine modulates energy storage. We showed that dopamine signaling regulates fat reservoirs in Caenorhabditis elegans. We found that the fat reducing effects of dopamine were dependent on dopaminergic receptors and a set of fat oxidation enzymes. Our findings reveal an ancient role for dopaminergic regulation of fat and suggest that dopamine signaling elicits this outcome through cascades that ultimately mobilize peripheral fat depots.
Collapse
Affiliation(s)
- Alexandre Guimarães de Almeida Barros
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jessika Cristina Bridi
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno Rezende de Souza
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Célio de Castro Júnior
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karen Cecília de Lima Torres
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leandro Malard
- Departamento de Física, Instituto de Ciências Exatas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ado Jorio
- Departamento de Física, Instituto de Ciências Exatas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora Marques de Miranda
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kaveh Ashrafi
- Department of Physiology, University of California San Francisco, San Francisco, California, United States
| | - Marco Aurélio Romano-Silva
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
212
|
Venkatachalam K, Luo J, Montell C. Evolutionarily conserved, multitasking TRP channels: lessons from worms and flies. Handb Exp Pharmacol 2014; 223:937-62. [PMID: 24961975 DOI: 10.1007/978-3-319-05161-1_9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The Transient Receptor Potential (TRP) channel family is comprised of a large group of cation-permeable channels, which display an extraordinary diversity of roles in sensory signaling. TRPs allow animals to detect chemicals, mechanical force, light, and changes in temperature. Consequently, these channels control a plethora of animal behaviors. Moreover, their functions are not limited to the classical senses, as they are cellular sensors, which are critical for ionic homeostasis and metabolism. Two genetically tractable invertebrate model organisms, Caenorhabditis elegans and Drosophila melanogaster, have led the way in revealing a wide array of sensory roles and behaviors that depend on TRP channels. Two overriding themes have emerged from these studies. First, TRPs are multitasking proteins, and second, many functions and modes of activation of these channels are evolutionarily conserved, including some that were formerly thought to be unique to invertebrates, such as phototransduction. Thus, worms and flies offer the potential to decipher roles for mammalian TRPs, which would otherwise not be suspected.
Collapse
Affiliation(s)
- Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, University of Texas School of Medicine, Houston, TX, 77030, USA,
| | | | | |
Collapse
|
213
|
Anderson A, McMullan R. From head to tail it's a two way street for neuro-immune communication. WORM 2014. [DOI: 10.4161/worm.29735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
214
|
Serotonergic chemosensory neurons modify the C. elegans immune response by regulating G-protein signaling in epithelial cells. PLoS Pathog 2013; 9:e1003787. [PMID: 24348250 PMCID: PMC3861540 DOI: 10.1371/journal.ppat.1003787] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/09/2013] [Indexed: 01/08/2023] Open
Abstract
The nervous and immune systems influence each other, allowing animals to rapidly protect themselves from changes in their internal and external environment. However, the complex nature of these systems in mammals makes it difficult to determine how neuronal signaling influences the immune response. Here we show that serotonin, synthesized in Caenorhabditis elegans chemosensory neurons, modulates the immune response. Serotonin released from these cells acts, directly or indirectly, to regulate G-protein signaling in epithelial cells. Signaling in these cells is required for the immune response to infection by the natural pathogen Microbacterium nematophilum. Here we show that serotonin signaling suppresses the innate immune response and limits the rate of pathogen clearance. We show that C. elegans uses classical neurotransmitters to alter the immune response. Serotonin released from sensory neurons may function to modify the immune system in response to changes in the animal's external environment such as the availability, or quality, of food.
Collapse
|
215
|
Zheng C, Karimzadegan S, Chiang V, Chalfie M. Histone methylation restrains the expression of subtype-specific genes during terminal neuronal differentiation in Caenorhabditis elegans. PLoS Genet 2013; 9:e1004017. [PMID: 24348272 PMCID: PMC3861114 DOI: 10.1371/journal.pgen.1004017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 10/25/2013] [Indexed: 01/16/2023] Open
Abstract
Although epigenetic control of stem cell fate choice is well established, little is known about epigenetic regulation of terminal neuronal differentiation. We found that some differences among the subtypes of Caenorhabditis elegans VC neurons, particularly the expression of the transcription factor gene unc-4, require histone modification, most likely H3K9 methylation. An EGF signal from the vulva alleviated the epigenetic repression of unc-4 in vulval VC neurons but not the more distant nonvulval VC cells, which kept unc-4 silenced. Loss of the H3K9 methyltransferase MET-2 or H3K9me2/3 binding proteins HPL-2 and LIN-61 or a novel chromodomain protein CEC-3 caused ectopic unc-4 expression in all VC neurons. Downstream of the EGF signaling in vulval VC neurons, the transcription factor LIN-11 and histone demethylases removed the suppressive histone marks and derepressed unc-4. Behaviorally, expression of UNC-4 in all the VC neurons caused an imbalance in the egg-laying circuit. Thus, epigenetic mechanisms help establish subtype-specific gene expression, which are needed for optimal activity of a neural circuit.
Collapse
Affiliation(s)
- Chaogu Zheng
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Siavash Karimzadegan
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Victor Chiang
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| |
Collapse
|
216
|
Noble T, Stieglitz J, Srinivasan S. An integrated serotonin and octopamine neuronal circuit directs the release of an endocrine signal to control C. elegans body fat. Cell Metab 2013; 18:672-84. [PMID: 24120942 PMCID: PMC3882029 DOI: 10.1016/j.cmet.2013.09.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 07/02/2013] [Accepted: 09/05/2013] [Indexed: 02/06/2023]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is an ancient and conserved neuromodulator of energy balance. Despite its importance, the neural circuits and molecular mechanisms underlying 5-HT-mediated control of body fat remain poorly understood. Here, we decipher the serotonergic neural circuit for body fat loss in C. elegans and show that the effects of 5-HT require signaling from octopamine, the invertebrate analog of adrenaline, to sustain body fat loss. Our results provide a potential molecular explanation for the long-observed potent effects of combined serotonergic and adrenergic weight loss drugs. In metabolic tissues, we find that the conserved regulatory adipocyte triglyceride lipase ATGL-1 drives serotonergic fat loss. We show that the serotonergic chloride channel MOD-1 relays a long-range endocrine signal from C. elegans body cavity neurons to control distal ATGL-1 function, via the nuclear receptor NHR-76. Our findings establish a conserved neuroendocrine axis operated by neural serotonergic and adrenergic-like signaling to regulate body fat.
Collapse
Affiliation(s)
- Tallie Noble
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jonathan Stieglitz
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Kellogg School of Science and Technology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Supriya Srinivasan
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
217
|
Serotonin control of thermotaxis memory behavior in nematode Caenorhabditis elegans. PLoS One 2013; 8:e77779. [PMID: 24223727 PMCID: PMC3815336 DOI: 10.1371/journal.pone.0077779] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/12/2013] [Indexed: 11/26/2022] Open
Abstract
Caenorhabditis elegans is as an ideal model system for the study of mechanisms underlying learning and memory. In the present study, we employed C. elegans assay system of thermotaxis memory to investigate the possible role of serotonin neurotransmitter in memory control. Our data showed that both mutations of tph-1, bas-1, and cat-4 genes, required for serotonin synthesis, and mutations of mod-5 gene, encoding a serotonin reuptake transporter, resulted in deficits in thermotaxis memory behavior. Exogenous treatment with serotonin effectively recovered the deficits in thermotaxis memory of tph-1 and bas-1 mutants to the level of wild-type N2. Neuron-specific activity assay of TPH-1 suggests that serotonin might regulate the thermotaxis memory behavior by release from the ADF sensory neurons. Ablation of ADF sensory neurons by expressing a cell-death activator gene egl-1 decreased the thermotaxis memory, whereas activation of ADF neurons by expression of a constitutively active protein kinase C homologue (pkc-1(gf)) increased the thermotaxis memory and rescued the deficits in thermotaxis memory in tph-1 mutants. Moreover, serotonin released from the ADF sensory neurons might act through the G-protein-coupled serotonin receptors of SER-4 and SER-7 to regulate the thermotaxis memory behavior. Genetic analysis implies that serotonin might further target the insulin signaling pathway to regulate the thermotaxis memory behavior. Thus, our results suggest the possible crucial role of serotonin and ADF sensory neurons in thermotaxis memory control in C. elegans.
Collapse
|
218
|
Soukas AA, Carr CE, Ruvkun G. Genetic regulation of Caenorhabditis elegans lysosome related organelle function. PLoS Genet 2013; 9:e1003908. [PMID: 24204312 PMCID: PMC3812091 DOI: 10.1371/journal.pgen.1003908] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 09/11/2013] [Indexed: 12/28/2022] Open
Abstract
Lysosomes are membrane-bound organelles that contain acid hydrolases that degrade cellular proteins, lipids, nucleic acids, and oligosaccharides, and are important for cellular maintenance and protection against age-related decline. Lysosome related organelles (LROs) are specialized lysosomes found in organisms from humans to worms, and share many of the features of classic lysosomes. Defective LROs are associated with human immune disorders and neurological disease. Caenorhabditis elegans LROs are the site of concentration of vital dyes such as Nile red as well as age-associated autofluorescence. Even though certain short-lived mutants have high LRO Nile red and high autofluorescence, and other long-lived mutants have low LRO Nile red and low autofluorescence, these two biologies are distinct. We identified a genetic pathway that modulates aging-related LRO phenotypes via serotonin signaling and the gene kat-1, which encodes a mitochondrial ketothiolase. Regulation of LRO phenotypes by serotonin and kat-1 in turn depend on the proton-coupled, transmembrane transporter SKAT-1. skat-1 loss of function mutations strongly suppress the high LRO Nile red accumulation phenotype of kat-1 mutation. Using a systems approach, we further analyzed the role of 571 genes in LRO biology. These results highlight a gene network that modulates LRO biology in a manner dependent upon the conserved protein kinase TOR complex 2. The results implicate new genetic pathways involved in LRO biology, aging related physiology, and potentially human diseases of the LRO. Lysosome related organelles (LROs) are specialized, membrane-bound organelles that share many common features of canonical lysosomes. Mutations in critical components of LRO biogenesis lead to human diseases of immunity, blood clotting, and pigmentation. In Caenorhabditis elegans, LROs are the site of accumulation of aging-related autofluorescence and the vital dye Nile red when fed to living C. elegans. Through classical genetics we show that the LRO is regulated by a conserved genetic pathway involving serotonin, a mitochondrial ketothiolase, and a proton-coupled solute transporter. Though previously thought to be linked in an obligatory manner, through systems level analysis we show that accumulation of C. elegans LRO Nile red and autofluorescence are mechanistically distinct processes. Contrary to the prior notion that LRO Nile red indicates lipid stores, we show that LRO Nile red is not correlated with, and may be anticorrelated with, C. elegans lipid stores. Using hundreds of candidate gene inactivations that disrupt Nile red accumulation, we determined which LRO regulatory genes specifically interact with 6 genetic mutants known to have altered LRO biology, identifying changes specifically dependent upon target of rapamycin complex 2 signaling. These data reveal relationships between LRO biology and aging and metabolism in C. elegans.
Collapse
Affiliation(s)
- Alexander A. Soukas
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (AAS); (GR)
| | - Christopher E. Carr
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Earth and Interplanetary Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Gary Ruvkun
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (AAS); (GR)
| |
Collapse
|
219
|
Migliarini S, Pacini G, Pelosi B, Lunardi G, Pasqualetti M. Lack of brain serotonin affects postnatal development and serotonergic neuronal circuitry formation. Mol Psychiatry 2013; 18:1106-18. [PMID: 23007167 DOI: 10.1038/mp.2012.128] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 12/11/2022]
Abstract
Despite increasing evidence suggests that serotonin (5-HT) can influence neurogenesis, neuronal migration and circuitry formation, the precise role of 5-HT on central nervous system (CNS) development is only beginning to be elucidated. Moreover, how changes in serotonin homeostasis during critical developmental periods may have etiological relevance to human mental disorders, remains an unsolved question. In this study we address the consequences of 5-HT synthesis abrogation on CNS development using a knock-in mouse line in which the tryptophan hydroxylase 2 (Tph2) gene is replaced by the eGFP reporter. We report that lack of brain 5-HT results in a dramatic reduction of body growth rate and in 60% lethality within the first 3 weeks after birth, with no gross anatomical changes in the brain. Thanks to the specific expression of the eGFP, we could highlight the serotonergic system independently of 5-HT immunoreactivity. We found that lack of central serotonin produces severe abnormalities in the serotonergic circuitry formation with a brain region- and time- specific effect. Indeed, we observed a striking reduction of serotonergic innervation to the suprachiasmatic and thalamic paraventricular nuclei, while a marked serotonergic hyperinnervation was found in the nucleus accumbens and hippocampus of Tph2∷eGFP mutants. Finally, we demonstrated that BDNF expression is significantly up-regulated in the hippocampus of mice lacking brain 5-HT, mirroring the timing of the appearance of hyperinnervation and thus unmasking a possible regulatory feedback mechanism tuning the serotonergic neuronal circuitry formation. On the whole, these findings reveal that alterations of serotonin levels during CNS development affect the proper wiring of the brain that may produce long-lasting changes leading to neurodevelopmental disorders.
Collapse
Affiliation(s)
- S Migliarini
- Department of Biology, Unit of Cellular and Developmental Biology, University of Pisa, Pisa, Italy
| | | | | | | | | |
Collapse
|
220
|
Song BM, Avery L. The pharynx of the nematode C. elegans: A model system for the study of motor control. WORM 2013; 2:e21833. [PMID: 24058858 PMCID: PMC3670459 DOI: 10.4161/worm.21833] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 08/14/2012] [Indexed: 11/19/2022]
Abstract
Motor control is a complex process that requires interplay among the nervous system, muscles and environment. The simple anatomy, well-characterized muscle movements and ample resources for molecular and cellular dissection make the pharynx of the nematode C. elegans an attractive model system for the study of motor control. The C. elegans pharynx shows two clear muscle movements that are essential for food intake, pharyngeal pumping and isthmus peristalsis. Here, we review our recent findings on the mechanism by which food activates the feeding motions. To understand this process, we characterized the behavior of the feeding motions in response to serotonin, an endogenous pharyngeal pumping activator whose action is triggered by food. We found that: (1) the timing of onset and frequencies of the two feeding motions are distinct; (2) isthmus peristalsis is selectively coupled to the preceding pump; (3) like food, serotonin activates isthmus peristalsis as well as pharyngeal pumping. By genetic analysis, we showed that two separate neural pathways activate the two feeding motions explaining the differences between the two feeding motions. We also proposed a model that explains how the two feeding motions are separately controlled, yet coupled by the interaction between the nervous system and the muscles in the pharynx. Finally, we briefly discuss future approaches to further understand the mechanism that couples the two feeding motions in C. elegans and to possibly understand evolution of motor control in the pharynx by expanding findings in C. elegans to other nematode species.
Collapse
Affiliation(s)
- Bo-Mi Song
- Department of Physiology and Biophysics; Virginia Commonwealth University; Richmond, VA USA
| | | |
Collapse
|
221
|
Schwarz J, Bringmann H. Reduced sleep-like quiescence in both hyperactive and hypoactive mutants of the Galphaq Gene egl-30 during lethargus in Caenorhabditis elegans. PLoS One 2013; 8:e75853. [PMID: 24073282 PMCID: PMC3779211 DOI: 10.1371/journal.pone.0075853] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/22/2013] [Indexed: 11/30/2022] Open
Abstract
Sleep-like states are characterized by massively reduced behavioral activity. Little is known about genetic control of sleep-like behavior. It is also not clear how general activity levels during wake-like behavior influence activity levels during sleep-like behavior. Mutations that increase wake-like activity are generally believed to also increase activity during sleep-like behavior and mutations that decrease wake-like activity are believed to have decreased activity during sleep-like behavior. We studied sleep-like behavior during lethargus in larvae of Caenorhabditis elegans. We looked through a small set of known mutants with altered activity levels. As expected, mutants with increased activity levels typically showed less sleep-like behavior. Among these hyperactive mutants was a gain-of-function mutant of the conserved heterotrimeric G protein subunit Galphaq gene egl-30. We found, however, that an unusual semidominant hypoactive mutant of egl-30 also had reduced sleep-like behavior. While movement was severely reduced and impaired in the semidominant egl-30 mutant, sleep-like behavior was severely reduced: the semidominant egl-30 mutant lacked prolonged periods of complete immobility, reduced spontaneous neural activity less, and reduced responsiveness to stimulation less. egl-30 is a well-known regulator of behavior. Our results suggest that egl-30 controls not only general activity levels, but also differences between wake-like and sleep-like behavior.
Collapse
Affiliation(s)
- Juliane Schwarz
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Henrik Bringmann
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- * E-mail:
| |
Collapse
|
222
|
Flavell SW, Pokala N, Macosko EZ, Albrecht DR, Larsch J, Bargmann CI. Serotonin and the neuropeptide PDF initiate and extend opposing behavioral states in C. elegans. Cell 2013; 154:1023-1035. [PMID: 23972393 PMCID: PMC3942133 DOI: 10.1016/j.cell.2013.08.001] [Citation(s) in RCA: 285] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 05/21/2013] [Accepted: 07/31/2013] [Indexed: 11/15/2022]
Abstract
Foraging animals have distinct exploration and exploitation behaviors that are organized into discrete behavioral states. Here, we characterize a neuromodulatory circuit that generates long-lasting roaming and dwelling states in Caenorhabditis elegans. We find that two opposing neuromodulators, serotonin and the neuropeptide pigment dispersing factor (PDF), each initiate and extend one behavioral state. Serotonin promotes dwelling states through the MOD-1 serotonin-gated chloride channel. The spontaneous activity of serotonergic neurons correlates with dwelling behavior, and optogenetic modulation of the critical MOD-1-expressing targets induces prolonged dwelling states. PDF promotes roaming states through a Gαs-coupled PDF receptor; optogenetic activation of cAMP production in PDF receptor-expressing cells induces prolonged roaming states. The neurons that produce and respond to each neuromodulator form a distributed circuit orthogonal to the classical wiring diagram, with several essential neurons that express each molecule. The slow temporal dynamics of this neuromodulatory circuit supplement fast motor circuits to organize long-lasting behavioral states.
Collapse
Affiliation(s)
- Steven W Flavell
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Navin Pokala
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Evan Z Macosko
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Dirk R Albrecht
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Johannes Larsch
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Cornelia I Bargmann
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
223
|
Pickett CL, Kornfeld K. Age-related degeneration of the egg-laying system promotes matricidal hatching in Caenorhabditis elegans. Aging Cell 2013; 12:544-53. [PMID: 23551912 PMCID: PMC4020343 DOI: 10.1111/acel.12079] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2013] [Indexed: 11/29/2022] Open
Abstract
The identification and characterization of age-related degenerative changes is a critical goal because it can elucidate mechanisms of aging biology and contribute to understanding interventions that promote longevity. Here, we document a novel, age-related degenerative change in C. elegans hermaphrodites, an important model system for the genetic analysis of longevity. Matricidal hatching--intra-uterine hatching of progeny that causes maternal death--displayed an age-related increase in frequency and affected ~70% of mated, wild-type hermaphrodites. The timing and incidence of matricidal hatching were largely independent of the levels of early and total progeny production and the duration of male exposure. Thus, matricidal hatching appears to reflect intrinsic age-related degeneration of the egg-laying system rather than use-dependent damage accumulation. Consistent with this model, mutations that extend longevity by causing dietary restriction significantly delayed matricidal hatching, indicating age-related degeneration of the egg-laying system is controlled by nutrient availability. To identify the underlying tissue defect, we analyzed serotonin signaling that triggers vulval muscle contractions. Mated hermaphrodites displayed an age-related decline in the ability to lay eggs in response to exogenous serotonin, indicating that vulval muscles and/or a further downstream function that is necessary for egg laying degenerate in an age-related manner. By characterizing a new, age-related degenerative event displayed by C. elegans hermaphrodites, these studies contribute to understanding a frequent cause of death in mated hermaphrodites and establish a model of age-related reproductive complications that may be relevant to the birthing process in other animals such as humans.
Collapse
Affiliation(s)
| | - Kerry Kornfeld
- Corresponding Author: Department of Developmental Biology, 660 South Euclid Ave., Campus Box 8103, Washington University School of Medicine, St. Louis, MO 63110, Telephone: (314) 747-1480, Fax: (314) 362-7058,
| |
Collapse
|
224
|
Lee H, Crane MM, Zhang Y, Lu H. Quantitative screening of genes regulating tryptophan hydroxylase transcription in Caenorhabditis elegans using microfluidics and an adaptive algorithm. Integr Biol (Camb) 2013; 5:372-80. [PMID: 23168494 DOI: 10.1039/c2ib20078c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Forward genetic screening via mutagenesis is a powerful method for identifying regulatory factors in target pathways in model organisms such as the soil-dwelling free-living nematode Caenorhabditis elegans (C. elegans). Currently manual microscopy is the standard technique for conducting such screens; however, it is labor-intensive and time-consuming because screening requires imaging thousands of animals. Recently microfluidic chips have been developed to increase the throughput of some of such experiments; nonetheless, most of these chips are multilayer devices and complicated to fabricate and therefore prone to failure during fabrication and operation. In addition, most sorting decisions are made manually and the criteria used for sorting are subjective. To overcome these limitations, we developed a simple single-layer microfluidic device and an adaptive algorithm to make sorting decisions. The one-layer device greatly improves the reliability, while quantitative analysis with the adaptive algorithm allows for the identification of mutations that generate subtle changes in expression, which would have been hard to detect by eye. The screening criterion is set based on the mutagenized population, not separate control populations measured prior to actual screening experiments, to account for stochasticity and day-to-day variations of gene expression in mutagenized worms. Moreover, during each experiment, the threshold is constantly updated to reflect the balance between maximizing sorting rate and minimizing false-positive rate. Using this system, we screened for mutants that have altered expression levels of tryptophan hydroxylase, a key enzyme for serotonin synthesis in a CaMKII gain-of-function background. We found several putative mutants in this screen. Furthermore, this microfluidic system and quantitative analysis can be easily adapted to study other pathways in C. elegans.
Collapse
Affiliation(s)
- Hyewon Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Dr NW, Atlanta, Georgia 30332, USA
| | | | | | | |
Collapse
|
225
|
Van Wielendaele P, Badisco L, Vanden Broeck J. Neuropeptidergic regulation of reproduction in insects. Gen Comp Endocrinol 2013; 188:23-34. [PMID: 23454669 DOI: 10.1016/j.ygcen.2013.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/01/2013] [Accepted: 02/10/2013] [Indexed: 12/17/2022]
Abstract
Successful animal reproduction depends on multiple physiological and behavioral processes that take place in a timely and orderly manner in both mating partners. It is not only necessary that all relevant processes are well coordinated, they also need to be adjusted to external factors of abiotic and biotic nature (e.g. population density, mating partner availability). Therefore, it is not surprising that several hormonal factors play a crucial role in the regulation of animal reproductive physiology. In insects (the largest class of animals on planet Earth), lipophilic hormones, such as ecdysteroids and juvenile hormones, as well as several neuropeptides take part in this complex regulation. While some peptides can affect reproduction via an indirect action (e.g. by influencing secretion of juvenile hormone), others exert their regulatory activity by directly targeting the reproductive system. In addition to insect peptides with proven activities, several others were suggested to also play a role in the regulation of reproductive physiology. Because of the long evolutionary history of many insect orders, it is not always clear to what extent functional data obtained in a given species can be extrapolated to other insect taxa. In this paper, we will review the current knowledge concerning the neuropeptidergic regulation of insect reproduction and situate it in a more general physiological context.
Collapse
Affiliation(s)
- Pieter Van Wielendaele
- Molecular Developmental Physiology and Signal Transduction, Department of Animal Physiology and Neurobiology, University of Leuven, Naamsestraat 59, P.O. Box 02465, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
226
|
Than MT, Kudlow BA, Han M. Functional analysis of neuronal microRNAs in Caenorhabditis elegans dauer formation by combinational genetics and Neuronal miRISC immunoprecipitation. PLoS Genet 2013; 9:e1003592. [PMID: 23818874 PMCID: PMC3688502 DOI: 10.1371/journal.pgen.1003592] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/09/2013] [Indexed: 01/15/2023] Open
Abstract
Identifying the physiological functions of microRNAs (miRNAs) is often challenging because miRNAs commonly impact gene expression under specific physiological conditions through complex miRNA::mRNA interaction networks and in coordination with other means of gene regulation, such as transcriptional regulation and protein degradation. Such complexity creates difficulties in dissecting miRNA functions through traditional genetic methods using individual miRNA mutations. To investigate the physiological functions of miRNAs in neurons, we combined a genetic “enhancer” approach complemented by biochemical analysis of neuronal miRNA-induced silencing complexes (miRISCs) in C. elegans. Total miRNA function can be compromised by mutating one of the two GW182 proteins (AIN-1), an important component of miRISC. We found that combining an ain-1 mutation with a mutation in unc-3, a neuronal transcription factor, resulted in an inappropriate entrance into the stress-induced, alternative larval stage known as dauer, indicating a role of miRNAs in preventing aberrant dauer formation. Analysis of this genetic interaction suggests that neuronal miRNAs perform such a role partly by regulating endogenous cyclic guanosine monophosphate (cGMP) signaling, potentially influencing two other dauer-regulating pathways. Through tissue-specific immunoprecipitations of miRISC, we identified miRNAs and their likely target mRNAs within neuronal tissue. We verified the biological relevance of several of these miRNAs and found that many miRNAs likely regulate dauer formation through multiple dauer-related targets. Further analysis of target mRNAs suggests potential miRNA involvement in various neuronal processes, but the importance of these miRNA::mRNA interactions remains unclear. Finally, we found that neuronal genes may be more highly regulated by miRNAs than intestinal genes. Overall, our study identifies miRNAs and their targets, and a physiological function of these miRNAs in neurons. It also suggests that compromising other aspects of gene expression, along with miRISC, can be an effective approach to reveal miRNA functions in specific tissues under specific physiological conditions. MicroRNAs (miRNAs) are important in the regulation of gene expression and are present in many organisms. To identify specific biological processes that are regulated by miRNAs, we disturbed total miRNA function under a certain genetic background and searched for defects. Interestingly, we found a prominent developmental defect that was dependent on a mutation in another gene involved in regulating transcription in neurons. Thus, by compromising two different aspects of gene regulation, we were able to identify a specific biological function of miRNAs. By investigating this defect, we determined that neuronal miRNAs likely function to help modulate cyclic guanosine monophosphate signaling. We then took a systematic approach and identified many miRNAs and genes that are likely to be regulated by neuronal miRNAs, and in doing so, we found genes involved in the initial defect. Additionally, we found many other genes, and show that genes expressed in neurons seem to be more regulated by miRNAs than genes in the intestine. Through our study, we identify a biological function of neuronal miRNAs and provide data that will help in identifying other important, novel, and exciting roles of this important class of small RNAs.
Collapse
Affiliation(s)
- Minh T Than
- Howard Hughes Medical Institute and Department of Molecular, Cellular, and Developmental Biology of University of Colorado, Boulder, Colorado, United States of America
| | | | | |
Collapse
|
227
|
Chen P, Martinez-Finley EJ, Bornhorst J, Chakraborty S, Aschner M. Metal-induced neurodegeneration in C. elegans. Front Aging Neurosci 2013; 5:18. [PMID: 23730287 PMCID: PMC3657624 DOI: 10.3389/fnagi.2013.00018] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/05/2013] [Indexed: 11/13/2022] Open
Abstract
The model species, Caenorhabditis elegans, has been used as a tool to probe for mechanisms underlying numerous neurodegenerative diseases. This use has been exploited to study neurodegeneration induced by metals. The allure of the nematode comes from the ease of genetic manipulation, the ability to fluorescently label neuronal subtypes, and the relative simplicity of the nervous system. Notably, C. elegans have approximately 60-80% of human genes and contain genes involved in metal homeostasis and transport, allowing for the study of metal-induced degeneration in the nematode. This review discusses methods to assess degeneration as well as outlines techniques for genetic manipulation and presents a comprehensive survey of the existing literature on metal-induced degeneration studies in the worm.
Collapse
Affiliation(s)
- Pan Chen
- Department of Pediatrics, Vanderbilt University Medical CenterNashville, TN, USA
| | | | - Julia Bornhorst
- Department of Pediatrics, Vanderbilt University Medical CenterNashville, TN, USA
| | - Sudipta Chakraborty
- Department of Pediatrics, Vanderbilt University Medical CenterNashville, TN, USA
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical CenterNashville, TN, USA
- Department of Pharmacology, the Kennedy Center for Research on Human Development, and the Center for Molecular Toxicology, Vanderbilt University Medical CenterNashville, TN, USA
| |
Collapse
|
228
|
Martorell P, Bataller E, Llopis S, Gonzalez N, Álvarez B, Montón F, Ortiz P, Ramón D, Genovés S. A cocoa peptide protects Caenorhabditis elegans from oxidative stress and β-amyloid peptide toxicity. PLoS One 2013; 8:e63283. [PMID: 23675471 PMCID: PMC3652819 DOI: 10.1371/journal.pone.0063283] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/29/2013] [Indexed: 11/18/2022] Open
Abstract
Background Cocoa and cocoa-based products contain different compounds with beneficial properties for human health. Polyphenols are the most frequently studied, and display antioxidant properties. Moreover, protein content is a very interesting source of antioxidant bioactive peptides, which can be used therapeutically for the prevention of age-related diseases. Methodology/Principal Findings A bioactive peptide, 13L (DNYDNSAGKWWVT), was obtained from a hydrolyzed cocoa by-product by chromatography. The in vitro inhibition of prolyl endopeptidase (PEP) was used as screening method to select the suitable fraction for peptide identification. Functional analysis of 13L peptide was achieved using the transgenic Caenorhabditis elegans strain CL4176 expressing the human Aβ1–42 peptide as a pre-clinical in vivo model for Alzheimer's disease. Among the peptides isolated, peptide 13L (1 µg/mL) showed the highest antioxidant activity (P≤0.001) in the wild-type strain (N2). Furthermore, 13L produced a significant delay in body paralysis in strain CL4176, especially in the 24–47 h period after Aβ1–42 peptide induction (P≤0.0001). This observation is in accordance with the reduction of Aβ deposits in CL4176 by western blot. Finally, transcriptomic analysis in wild-type nematodes treated with 13L revealed modulation of the proteosomal and synaptic functions as the main metabolic targets of the peptide. Conclusions/Significance These findings suggest that the cocoa 13L peptide has antioxidant activity and may reduce Aβ deposition in a C. elegans model of Alzheimer's disease; and therefore has a putative therapeutic potential for prevention of age-related diseases. Further studies in murine models and humans will be essential to analyze the effectiveness of the 13L peptide in higher animals.
Collapse
Affiliation(s)
- Patricia Martorell
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Esther Bataller
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Silvia Llopis
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Núria Gonzalez
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Beatriz Álvarez
- Molecular Biology Laboratory, Microbial Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Fernando Montón
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Pepa Ortiz
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Daniel Ramón
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
| | - Salvador Genovés
- Cell Biology Laboratory, Food Biotechnology Department, Biópolis SL, Paterna, Valencia, Spain
- * E-mail:
| |
Collapse
|
229
|
Weinberg P, Flames N, Sawa H, Garriga G, Hobert O. The SWI/SNF chromatin remodeling complex selectively affects multiple aspects of serotonergic neuron differentiation. Genetics 2013; 194:189-98. [PMID: 23457234 PMCID: PMC3632466 DOI: 10.1534/genetics.112.148742] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/25/2013] [Indexed: 01/07/2023] Open
Abstract
Regulatory programs that control the specification of serotonergic neurons have been investigated by genetic mutant screens in the nematode Caenorhabditis elegans. Loss of a previously uncloned gene, ham-3, affects migration and serotonin antibody staining of the hermaphrodite-specific neuron (HSN) pair. We characterize these defects here in more detail, showing that the defects in serotonin antibody staining are paralleled by a loss of the transcription of all genes involved in serotonin synthesis and transport. This loss is specific to the HSN class as other serotonergic neurons appear to differentiate normally in ham-3 null mutants. Besides failing to migrate appropriately, the HSNs also display axon pathfinding defects in ham-3 mutants. However, the HSNs are still generated and express a subset of their terminal differentiation features in ham-3 null mutants, demonstrating that ham-3 is a specific regulator of select features of the HSNs. We show that ham-3 codes for the C. elegans ortholog of human BAF60, Drosophila Bap60, and yeast Swp73/Rsc6, which are subunits of the yeast SWI/SNF and vertebrate BAF chromatin remodeling complex. We show that the effect of ham-3 on serotonergic fate can be explained by ham-3 regulating the expression of the Spalt/SALL-type Zn finger transcription factor sem-4, a previously identified regulator of serotonin expression in HSNs and of the ham-2 Zn transcription factor, a previously identified regulator of HSN migration and axon outgrowth. Our findings provide the first evidence for the involvement of the BAF complex in the acquisition of terminal neuronal identity and constitute genetic proof by germline knockout that a BAF complex component can have cell-type-specific roles during development.
Collapse
Affiliation(s)
- Peter Weinberg
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032
| | - Nuria Flames
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, E-46010 Valencia, Spain
| | - Hitoshi Sawa
- National Institute of Genetics, 411-8540 Mishima, Japan
| | - Gian Garriga
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200
| | - Oliver Hobert
- Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
230
|
Chen Z, Hendricks M, Cornils A, Maier W, Alcedo J, Zhang Y. Two insulin-like peptides antagonistically regulate aversive olfactory learning in C. elegans. Neuron 2013; 77:572-85. [PMID: 23395381 DOI: 10.1016/j.neuron.2012.11.025] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2012] [Indexed: 12/11/2022]
Abstract
The insulin/insulin-like peptides (ILPs) regulate key events in physiology, including neural plasticity. However, the cellular and circuit mechanisms whereby ILPs regulate learning remain largely unknown. Here, we characterize two ILPs that play antagonistic roles in aversive olfactory learning of C. elegans. We show that the ILP ins-6 acts from ASI sensory neurons to enable learning by repressing the transcription of another ILP, ins-7, specifically in URX neurons. A high level of INS-7 from URX disrupts learning by antagonizing the insulin receptor-like homolog DAF-2 in the postsynaptic neurons RIA, which play an essential role in the neural circuit underlying olfactory learning. We also show that increasing URX-generated INS-7 and loss of INS-6, both of which abolish learning, alter RIA neuronal property. Together, our results reveal an "ILP-to-ILP" pathway that links environment-sensing neurons, ASI and URX, to the key neuron, RIA, of a network that underlies olfactory plasticity and modulates its activity.
Collapse
Affiliation(s)
- Zhunan Chen
- Department of Organismic and Evolutionary Biology, The Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | |
Collapse
|
231
|
Hashmi S, Wang Y, Parhar RS, Collison KS, Conca W, Al-Mohanna F, Gaugler R. A C. elegans model to study human metabolic regulation. Nutr Metab (Lond) 2013; 10:31. [PMID: 23557393 PMCID: PMC3636097 DOI: 10.1186/1743-7075-10-31] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/19/2013] [Indexed: 12/16/2022] Open
Abstract
Lipid metabolic disorder is a critical risk factor for metabolic syndrome, triggering debilitating diseases like obesity and diabetes. Both obesity and diabetes are the epicenter of important medical issues, representing a major international public health threat. Accumulation of fat in adipose tissue, muscles and liver and/or the defects in their ability to metabolize fatty acids, results in insulin resistance. This triggers an early pathogenesis of type 2 diabetes (T2D). In mammals, lipid metabolism involves several organs, including the brain, adipose tissue, muscles, liver, and gut. These organs are part of complex homeostatic system and communicate through hormones, neurons and metabolites. Our study dissects the importance of mammalian Krüppel-like factors in over all energy homeostasis. Factors controlling energy metabolism are conserved between mammals and Caenorhabditis elegans providing a new and powerful strategy to delineate the molecular pathways that lead to metabolic disorder. The C. elegans intestine is our model system where genetics, molecular biology, and cell biology are used to identify and understand genes required in fat metabolism. Thus far, we have found an important role of C. elegans KLF in FA biosynthesis, mitochondrial proliferation, lipid secretion, and β-oxidation. The mechanism by which KLF controls these events in lipid metabolism is unknown. We have recently observed that C. elegans KLF-3 selectively acts on insulin components to regulate insulin pathway activity. There are many factors that control energy homeostasis and defects in this control system are implicated in the pathogenesis of human obesity and diabetes. In this review we are discussing a role of KLF in human metabolic regulation.
Collapse
Affiliation(s)
- Sarwar Hashmi
- Laboratory of Developmental Biology, Center for Vector Biology, Rutgers University, 180 Jones Avenue, New Brunswick, NJ, 08901, USA.
| | | | | | | | | | | | | |
Collapse
|
232
|
A neuronal signaling pathway of CaMKII and Gqα regulates experience-dependent transcription of tph-1. J Neurosci 2013; 33:925-35. [PMID: 23325232 DOI: 10.1523/jneurosci.2355-12.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Dynamic serotonin biosynthesis is important for serotonin function; however, the mechanisms that underlie experience-dependent transcriptional regulation of the rate-limiting serotonin biosynthetic enzyme tryptophan hydroxylase (TPH) are poorly understood. Here, we characterize the molecular and cellular mechanisms that regulate increased transcription of Caenorhabditis elegans tph-1 in a pair of serotonergic neurons ADF during an aversive experience with pathogenic bacteria, a common environmental peril for worms. Training with pathogenic bacteria induces a learned aversion to the smell of the pathogen, a behavioral plasticity that depends on the serotonin signal from ADF neurons. We demonstrate that pathogen training increases ADF neuronal activity. While activating ADF increases tph-1 transcription, inhibiting ADF activity abolishes the training effect on tph-1, demonstrating the dependence of tph-1 transcriptional regulation on ADF neural activity. At the molecular level, the C. elegans homolog of CaMKII, UNC-43, functions cell-autonomously in ADF neurons to generate training-dependent enhancement in neuronal activity and tph-1 transcription, and this cell-autonomous function of UNC-43 is required for learning. Furthermore, selective expression of an activated form of UNC-43 in ADF neurons is sufficient to increase ADF activity and tph-1 transcription, mimicking the training effect. Upstream of ADF, the Gqα protein EGL-30 facilitates training-dependent induction of tph-1 by functional regulation of olfactory sensory neurons, which underscores the importance of sensory experience. Together, our work elucidates the molecular and cellular mechanisms whereby experience modulates tph-1 transcription.
Collapse
|
233
|
Xie Y, Moussaif M, Choi S, Xu L, Sze JY. RFX transcription factor DAF-19 regulates 5-HT and innate immune responses to pathogenic bacteria in Caenorhabditis elegans. PLoS Genet 2013; 9:e1003324. [PMID: 23505381 PMCID: PMC3591283 DOI: 10.1371/journal.pgen.1003324] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 01/04/2013] [Indexed: 02/06/2023] Open
Abstract
In Caenorhabditis elegans the Toll-interleukin receptor domain adaptor protein TIR-1 via a conserved mitogen-activated protein kinase (MAPK) signaling cascade induces innate immunity and upregulates serotonin (5-HT) biosynthesis gene tph-1 in a pair of ADF chemosensory neurons in response to infection. Here, we identify transcription factors downstream of the TIR-1 signaling pathway. We show that common transcription factors control the innate immunity and 5-HT biosynthesis. We demonstrate that a cysteine to tyrosine substitution in an ARM motif of the HEAT/Arm repeat region of the TIR-1 protein confers TIR-1 hyperactivation, leading to constitutive tph-1 upregulation in the ADF neurons, increased expression of intestinal antimicrobial genes, and enhanced resistance to killing by the human opportunistic pathogen Pseudomonas aeruginosa PA14. A forward genetic screen for suppressors of the hyperactive TIR-1 led to the identification of DAF-19, an ortholog of regulatory factor X (RFX) transcription factors that are required for human adaptive immunity. We show that DAF-19 concerts with ATF-7, a member of the activating transcription factor (ATF)/cAMP response element-binding B (CREB) family of transcription factors, to regulate tph-1 and antimicrobial genes, reminiscent of RFX-CREB interaction in human immune cells. daf-19 mutants display heightened susceptibility to killing by PA14. Remarkably, whereas the TIR-1-MAPK-DAF-19/ATF-7 pathway in the intestinal immunity is regulated by DKF-2/protein kinase D, we found that the regulation of tph-1 expression is independent of DKF-2 but requires UNC-43/Ca2+/calmodulin-dependent protein kinase (CaMK) II. Our results suggest that pathogenic cues trigger a common core-signaling pathway via tissue-specific mechanisms and demonstrate a novel role for RFX factors in neuronal and innate immune responses to infection. Toll-interleukin receptor (TIR)–domain adaptor proteins are keys to activate signaling cascades inducing transcriptional responses to internal and external pathogenic signals in evolutionary disparate organisms. Despite lacking a homolog of the mammalian innate immunity transcriptional regulator nuclear factor-kappaB (NF-κB), the nematode Caenorhabditis elegans responds to infections by activating TIR-1 signaling targets in the innate immune system and in neurons. Through a genetic screen for factors required for TIR-1 signaling to upregulate the serotonin biosynthesis gene tph-1, we identified DAF-19, an ortholog of regulatory factor X (RFX) transcription factors that were initially discovered in human immune cells. We show that DAF-19 concerts with ATF-7, a member of the activating transcription factor (ATF)/cAMP response element-binding B (CREB) family of transcription factors, to upregulate tph-1 in the ADF chemosensory neurons and antimicrobial genes in the intestine in response to bacterial infection, reminiscent of RFX-CREB interaction in human immune cells. daf-19 mutants display heightened susceptibility to killing by the human pathogen Pseudomonas aeruginosa PA14. Our studies suggest that RFX transcriptional regulation, which is essential for human adaptive immunity, has an ancient role in controlling serotonin biosynthesis and innate immunity.
Collapse
Affiliation(s)
| | | | | | | | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
234
|
Song BM, Faumont S, Lockery S, Avery L. Recognition of familiar food activates feeding via an endocrine serotonin signal in Caenorhabditis elegans. eLife 2013; 2:e00329. [PMID: 23390589 PMCID: PMC3564447 DOI: 10.7554/elife.00329] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/22/2012] [Indexed: 02/04/2023] Open
Abstract
Familiarity discrimination has a significant impact on the pattern of food intake across species. However, the mechanism by which the recognition memory controls feeding is unclear. Here, we show that the nematode Caenorhabditis elegans forms a memory of particular foods after experience and displays behavioral plasticity, increasing the feeding response when they subsequently recognize the familiar food. We found that recognition of familiar food activates the pair of ADF chemosensory neurons, which subsequently increase serotonin release. The released serotonin activates the feeding response mainly by acting humorally and directly activates SER-7, a type 7 serotonin receptor, in MC motor neurons in the feeding organ. Our data suggest that worms sense the taste and/or smell of novel bacteria, which overrides the stimulatory effect of familiar bacteria on feeding by suppressing the activity of ADF or its upstream neurons. Our study provides insight into the mechanism by which familiarity discrimination alters behavior.DOI:http://dx.doi.org/10.7554/eLife.00329.001.
Collapse
Affiliation(s)
- Bo-mi Song
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, United States
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Serge Faumont
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Shawn Lockery
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Leon Avery
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, United States
| |
Collapse
|
235
|
Schumacher JA, Hsieh YW, Chen S, Pirri JK, Alkema MJ, Li WH, Chang C, Chuang CF. Intercellular calcium signaling in a gap junction-coupled cell network establishes asymmetric neuronal fates in C. elegans. Development 2013; 139:4191-201. [PMID: 23093425 DOI: 10.1242/dev.083428] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The C. elegans left and right AWC olfactory neurons specify asymmetric subtypes, one default AWC(OFF) and one induced AWC(ON), through a stochastic, coordinated cell signaling event. Intercellular communication between AWCs and non-AWC neurons via a NSY-5 gap junction network coordinates AWC asymmetry. However, the nature of intercellular signaling across the network and how individual non-AWC cells in the network influence AWC asymmetry is not known. Here, we demonstrate that intercellular calcium signaling through the NSY-5 gap junction neural network coordinates a precise 1AWC(ON)/1AWC(OFF) decision. We show that NSY-5 gap junctions in C. elegans cells mediate small molecule passage. We expressed vertebrate calcium-buffer proteins in groups of cells in the network to reduce intracellular calcium levels, thereby disrupting intercellular communication. We find that calcium in non-AWC cells of the network promotes the AWC(ON) fate, in contrast to the autonomous role of calcium in AWCs to promote the AWC(OFF) fate. In addition, calcium in specific non-AWCs promotes AWC(ON) side biases through NSY-5 gap junctions. Our results suggest a novel model in which calcium has dual roles within the NSY-5 network: autonomously promoting AWC(OFF) and non-autonomously promoting AWC(ON).
Collapse
Affiliation(s)
- Jennifer A Schumacher
- Division of Developmental Biology, Children's Hospital Medical Center Research Foundation, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
236
|
Menon R, Gasser RB, Mitreva M, Ranganathan S. An analysis of the transcriptome of Teladorsagia circumcincta: its biological and biotechnological implications. BMC Genomics 2012; 13 Suppl 7:S10. [PMID: 23282110 PMCID: PMC3521389 DOI: 10.1186/1471-2164-13-s7-s10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Teladorsagia circumcincta (order Strongylida) is an economically important parasitic nematode of small ruminants (including sheep and goats) in temperate climatic regions of the world. Improved insights into the molecular biology of this parasite could underpin alternative methods required to control this and related parasites, in order to circumvent major problems associated with anthelmintic resistance. The aims of the present study were to define the transcriptome of the adult stage of T. circumcincta and to infer the main pathways linked to molecules known to be expressed in this nematode. Since sheep develop acquired immunity against T. circumcincta, there is some potential for the development of a vaccine against this parasite. Hence, we infer excretory/secretory molecules for T. circumcincta as possible immunogens and vaccine candidates. RESULTS A total of 407,357 ESTs were assembled yielding 39,852 putative gene sequences. Conceptual translation predicted 24,013 proteins, which were then subjected to detailed annotation which included pathway mapping of predicted proteins (including 112 excreted/secreted [ES] and 226 transmembrane peptides), domain analysis and GO annotation was carried out using InterProScan along with BLAST2GO. Further analysis was carried out for secretory signal peptides using SignalP and non-classical sec pathway using SecretomeP tools. For ES proteins, key pathways, including Fc epsilon RI, T cell receptor, and chemokine signalling as well as leukocyte transendothelial migration were inferred to be linked to immune responses, along with other pathways related to neurodegenerative diseases and infectious diseases, which warrant detailed future studies. KAAS could identify new and updated pathways like phagosome and protein processing in endoplasmic reticulum. Domain analysis for the assembled dataset revealed families of serine, cysteine and proteinase inhibitors which might represent targets for parasite intervention. InterProScan could identify GO terms pertaining to the extracellular region. Some of the important domain families identified included the SCP-like extracellular proteins which belong to the pathogenesis-related proteins (PRPs) superfamily along with C-type lectin, saposin-like proteins. The 'extracellular region' that corresponds to allergen V5/Tpx-1 related, considered important in parasite-host interactions, was also identified. Six cysteine motif (SXC1) proteins, transthyretin proteins, C-type lectins, activation-associated secreted proteins (ASPs), which could represent potential candidates for developing novel anthelmintics or vaccines were few other important findings. Of these, SXC1, protein kinase domain-containing protein, trypsin family protein, trypsin-like protease family member (TRY-1), putative major allergen and putative lipid binding protein were identified which have not been reported in the published T. circumcincta proteomics analysis. Detailed analysis of 6,058 raw EST sequences from dbEST revealed 315 putatively secreted proteins. Amongst them, C-type single domain activation associated secreted protein ASP3 precursor, activation-associated secreted proteins (ASP-like protein), cathepsin B-like cysteine protease, cathepsin L cysteine protease, cysteine protease, TransThyretin-Related and Venom-Allergen-like proteins were the key findings. CONCLUSIONS We have annotated a large dataset ESTs of T. circumcincta and undertaken detailed comparative bioinformatics analyses. The results provide a comprehensive insight into the molecular biology of this parasite and disease manifestation which provides potential focal point for future research. We identified a number of pathways responsible for immune response. This type of large-scale computational scanning could be coupled with proteomic and metabolomic studies of this parasite leading to novel therapeutic intervention and disease control strategies. We have also successfully affirmed the use of bioinformatics tools, for the study of ESTs, which could now serve as a benchmark for the development of new computational EST analysis pipelines.
Collapse
Affiliation(s)
- Ranjeeta Menon
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | | | | | | |
Collapse
|
237
|
Curran KP, Chalasani SH. Serotonin circuits and anxiety: what can invertebrates teach us? INVERTEBRATE NEUROSCIENCE : IN 2012; 12:81-92. [PMID: 22918570 PMCID: PMC3505513 DOI: 10.1007/s10158-012-0140-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/26/2012] [Indexed: 11/08/2022]
Abstract
Fear, a reaction to a threatening situation, is a broadly adaptive feature crucial to the survival and reproductive fitness of individual organisms. By contrast, anxiety is an inappropriate behavioral response often to a perceived, not real, threat. Functional imaging, biochemical analysis, and lesion studies with humans have identified the HPA axis and the amygdala as key neuroanatomical regions driving both fear and anxiety. Abnormalities in these biological systems lead to misregulated fear and anxiety behaviors such as panic attacks and post-traumatic stress disorders. These behaviors are often treated by increasing serotonin levels at synapses, suggesting a role for serotonin signaling in ameliorating both fear and anxiety. Interestingly, serotonin signaling is highly conserved between mammals and invertebrates. We propose that genetically tractable invertebrate models organisms, such as Drosophila melanogaster and Caenorhabditis elegans, are ideally suited to unravel the complexity of the serotonin signaling pathways. These model systems possess well-defined neuroanatomies and robust serotonin-mediated behavior and should reveal insights into how serotonin can modulate human cognitive functions.
Collapse
Affiliation(s)
- Kevin P. Curran
- Molecular Neurobiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037 USA
| | - Sreekanth H. Chalasani
- Molecular Neurobiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
238
|
Gürel G, Gustafson MA, Pepper JS, Horvitz HR, Koelle MR. Receptors and other signaling proteins required for serotonin control of locomotion in Caenorhabditis elegans. Genetics 2012; 192:1359-71. [PMID: 23023001 PMCID: PMC3512144 DOI: 10.1534/genetics.112.142125] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/15/2012] [Indexed: 01/05/2023] Open
Abstract
A better understanding of the molecular mechanisms of signaling by the neurotransmitter serotonin is required to assess the hypothesis that defects in serotonin signaling underlie depression in humans. Caenorhabditis elegans uses serotonin as a neurotransmitter to regulate locomotion, providing a genetic system to analyze serotonin signaling. From large-scale genetic screens we identified 36 mutants of C. elegans in which serotonin fails to have its normal effect of slowing locomotion, and we molecularly identified eight genes affected by 19 of the mutations. Two of the genes encode the serotonin-gated ion channel MOD-1 and the G-protein-coupled serotonin receptor SER-4. mod-1 is expressed in the neurons and muscles that directly control locomotion, while ser-4 is expressed in an almost entirely non-overlapping set of sensory and interneurons. The cells expressing the two receptors are largely not direct postsynaptic targets of serotonergic neurons. We analyzed animals lacking or overexpressing the receptors in various combinations using several assays for serotonin response. We found that the two receptors act in parallel to affect locomotion. Our results show that serotonin functions as an extrasynaptic signal that independently activates multiple receptors at a distance from its release sites and identify at least six additional proteins that appear to act with serotonin receptors to mediate serotonin response.
Collapse
Affiliation(s)
- Güliz Gürel
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Megan A. Gustafson
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Judy S. Pepper
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| | - H. Robert Horvitz
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Michael R. Koelle
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
239
|
Palgunow D, Klapper M, Döring F. Dietary restriction during development enlarges intestinal and hypodermal lipid droplets in Caenorhabditis elegans. PLoS One 2012. [PMID: 23185233 PMCID: PMC3502458 DOI: 10.1371/journal.pone.0046198] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dietary restriction (DR) extends lifespan in man species and modulates evolutionary conserved signalling and metabolic pathways. Most of these studies were done in adult animals. Here we investigated fat phenotypes of C. elegans larvae and adults which were exposed to DR during development. This approach was named "developmental-DR" (dDR). Moderate as well as stringent dDR increased the triglyceride to protein ratio in L4 larvae and adult worms. This alteration was accompanied by a marked expansion of intestinal and hypodermal lipid droplets. In comparison to ad libitum condition, the relative proportion of fat stored in large lipid droplets (>50 µm(3)) was increased by a factor of about 5 to 6 in larvae exposed to dDR. Microarray-based expression profiling identified several dDR-regulated genes of lipolysis and lipogenesis which may contribute to the observed fat phenotypes. In conclusion, dDR increases the triglyceride to protein ratio, enlarges lipid droplets and alters the expression of genes functioning in lipid metabolism in C. elegans. These changes might be an effective adaptation to conserve fat stores in animals subjected to limiting food supply during development.
Collapse
Affiliation(s)
| | | | - Frank Döring
- Department of Molecular Prevention, Institute of Human Nutrition and Food Science, Christian-Albrechts-University of Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
240
|
Osanai A, Hu DL, Nakane A. Caenorhabditis elegansavoids staphylococcal superantigenic toxins via 5-hydroxytryptamine-dependent pathway. Can J Microbiol 2012; 58:1268-77. [DOI: 10.1139/w2012-107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Avoidance behavior of Caenorhabditis elegans, a nematode, towards Staphylococcus aureus, a pathogenic bacterium, was studied. Caenorhabditis elegans avoided S. aureus cultures and also their culture supernatants, suggesting that secretory molecules are involved in the repellent activity. We demonstrated that toxic shock syndrome toxin 1 (TSST-1) and staphylococcal enterotoxin C (SEC), the superantigenic toxins produced by S. aureus, are responsible for the nematode avoidance. By using TSST-1 and SEC mutants, the results indicated that the repellent activity of these toxins is independent of their superantigenic activity. The TSST-1 and SEC were found to locate at chemosensory neurons that are responsible for the recognition of repellents and avoidance of pathogenic bacteria. When mutants of C. elegans deficient in Toll/interleukin-1 receptor (TIR-1) and 5-hydroxytryptamine (5-HT) biosynthesis were used, avoidance behavior was attenuated. In the 5-HT biosynthesis deficient mutant nematodes, the avoidance activity was recovered when exogenous 5-HT was added. tph-1 expression and 5-HT production were upregulated when the nematodes were treated with TSST-1 or SEC. These results suggest that C. elegans avoids S. aureus by recognizing secretory molecules including TSST-1 and SEC and this avoidance is dependent on TIR and production of 5-HT.
Collapse
Affiliation(s)
- Arihiro Osanai
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, 036-8562, Japan
| | - Dong-Liang Hu
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, 036-8562, Japan
| | - Akio Nakane
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
241
|
Serotonin: from top to bottom. Biogerontology 2012; 14:21-45. [PMID: 23100172 DOI: 10.1007/s10522-012-9406-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/08/2012] [Indexed: 01/13/2023]
Abstract
Serotonin is a monoamine neurotransmitter, which is phylogenetically conserved in a wide range of species from nematodes to humans. In mammals, age-related changes in serotonin systems are known risk factors of age-related diseases, such as diabetes, faecal incontinence and cardiovascular diseases. A decline in serotonin function with aging would be consistent with observations of age-related changes in behaviours, such as sleep, sexual behaviour and mood all of which are linked to serotonergic function. Despite this little is known about serotonin in relation to aging. This review aims to give a comprehensive analysis of the distribution, function and interactions of serotonin in the brain; gastrointestinal tract; skeletal; vascular and immune systems. It also aims to demonstrate how the function of serotonin is linked to aging and disease pathology in these systems. The regulation of serotonin via microRNAs is also discussed, as are possible applications of serotonergic drugs in aging research and age-related diseases. Furthermore, this review demonstrates that serotonin is potentially involved in whole organism aging through its links with multiple organs, the immune system and microRNA regulation. Methods to investigate these links are discussed.
Collapse
|
242
|
Antebi A. Regulation of longevity by the reproductive system. Exp Gerontol 2012; 48:596-602. [PMID: 23063987 DOI: 10.1016/j.exger.2012.09.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 09/06/2012] [Accepted: 09/20/2012] [Indexed: 01/04/2023]
Abstract
Pioneering work in model organisms reveals that the reproductive system is involved not only in propagation of the species but also regulates organismal metabolism and longevity. In C. elegans, prevention of germline stem cell proliferation results in a 60% extension of lifespan, termed gonadal longevity. Gonadal longevity relies on the transcriptional activities of steroid nuclear receptor DAF-12, the FOXO transcription factor homolog DAF-16, the FOXA transcription factor homolog PHA-4, and the HNF-4-like nuclear receptor NHR-80. These transcription factors work in an integrated transcriptional network to regulate fatty acid lipolysis, autophagy, stress resistance and other processes, which altogether enhance homeostasis and extend life. Because the reproductive system also regulates longevity in other species, studies in C. elegans may shed light on ancient mechanisms governing reproduction and survival.
Collapse
Affiliation(s)
- Adam Antebi
- Max Planck Institute for Biology of Ageing, Gleueler Str. 50a, D-50931 Cologne, Germany.
| |
Collapse
|
243
|
Okazaki A, Takagi S. An optogenetic application of proton pump ArchT to C. elegans cells. Neurosci Res 2012; 75:29-34. [PMID: 23044183 DOI: 10.1016/j.neures.2012.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/20/2012] [Accepted: 08/30/2012] [Indexed: 11/27/2022]
Abstract
Application of novel light-driven ion channel/pumps would benefit optogenetic studies of Caenorhabditis elegans. A recent study showed that ArchT, a novel light-driven outward proton pump, is >3 times more light-sensitive than the Arch proton pump. Here we report the silencing effect of ArchT in C. elegans cells. ArchT expressed by using a body-wall muscle or pan-neuronal-promoters caused a quick and reliable locomotion paralysis when worms were illuminated by green light. Unlike the report on mouse neurons, however, light sensitivity of ArchT is similar to that of Arch in C. elegans. ArchT-mediated acute silencing of serotonergic neurons quickly triggered backward locomotion. This response was abolished in the presence of exogenously added serotonin, suggesting that, in a normal situation, serotonin is secreted in a constitutive fashion to repress backward movement.
Collapse
Affiliation(s)
- Ayako Okazaki
- Division of Biological Science, Nagoya University Graduate School of Science, Chikusa-ku, Nagoya 464-8602, Japan
| | | |
Collapse
|
244
|
Abstract
Food is important to any animal, and a large part of the behavioral repertoire is concerned with ensuring adequate nutrition. Two main nutritional sensations, hunger and satiety, produce opposite behaviors. Hungry animals seek food, increase exploratory behavior and continue feeding once they encounter food. Satiated animals decrease exploratory behavior, take rest, and stop feeding. The signals of hunger or satiety and their effects on physiology and behavior will depend not only on the animal's current nutritional status but also on its experience and the environment in which the animal evolved. In our novel, nutritionally rich environment, improper control of appetite contributes to diseases from anorexia to the current epidemic of obesity. Despite extraordinary recent advances, genetic contribution to appetite control is still poorly understood partly due to lack of simple genetic model systems. In this review, we will discuss current understanding of molecular and cellular mechanisms by which animals regulate food intake depending on their nutritional status. Then, focusing on relatively less known muscarinic and cGMP signals, we will discuss how the molecular and behavioral aspects of hunger and satiety are conserved in a simple invertebrate model system, C. elegans so as for us to use it to understand the genetics of appetite control.
Collapse
Affiliation(s)
- Young-Jai You
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | |
Collapse
|
245
|
Delaying aging and the aging-associated decline in protein homeostasis by inhibition of tryptophan degradation. Proc Natl Acad Sci U S A 2012; 109:14912-7. [PMID: 22927396 DOI: 10.1073/pnas.1203083109] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Toxicity of aggregation-prone proteins is thought to play an important role in aging and age-related neurological diseases like Parkinson and Alzheimer's diseases. Here, we identify tryptophan 2,3-dioxygenase (tdo-2), the first enzyme in the kynurenine pathway of tryptophan degradation, as a metabolic regulator of age-related α-synuclein toxicity in a Caenorhabditis elegans model. Depletion of tdo-2 also suppresses toxicity of other heterologous aggregation-prone proteins, including amyloid-β and polyglutamine proteins, and endogenous metastable proteins that are sensors of normal protein homeostasis. This finding suggests that tdo-2 functions as a general regulator of protein homeostasis. Analysis of metabolite levels in C. elegans strains with mutations in enzymes that act downstream of tdo-2 indicates that this suppression of toxicity is independent of downstream metabolites in the kynurenine pathway. Depletion of tdo-2 increases tryptophan levels, and feeding worms with extra L-tryptophan also suppresses toxicity, suggesting that tdo-2 regulates proteotoxicity through tryptophan. Depletion of tdo-2 extends lifespan in these worms. Together, these results implicate tdo-2 as a metabolic switch of age-related protein homeostasis and lifespan. With TDO and Indoleamine 2,3-dioxygenase as evolutionarily conserved human orthologs of TDO-2, intervening with tryptophan metabolism may offer avenues to reducing proteotoxicity in aging and age-related diseases.
Collapse
|
246
|
Nishio N, Mohri-Shiomi A, Nishida Y, Hiramatsu N, Kodama-Namba E, Kimura KD, Kuhara A, Mori I. A novel and conserved protein AHO-3 is required for thermotactic plasticity associated with feeding states in Caenorhabditis elegans. Genes Cells 2012; 17:365-86. [PMID: 22512337 PMCID: PMC3506735 DOI: 10.1111/j.1365-2443.2012.01594.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although a large proportion of molecules expressed in the nervous system are conserved from invertebrate to vertebrate, functional properties of such molecules are less characterized. Here, we show that highly conserved hydrolase AHO-3 acts as a novel regulator of starvation-induced thermotactic plasticity in Caenorhabditis elegans. As wild-type animals, aho-3 mutants migrated to the cultivation temperature on a linear thermal gradient after cultivation at a particular temperature with food. Whereas wild-type animals cultivated under food-deprived condition showed dispersed distribution on the gradient, aho-3 mutants exhibited tendency to migrate toward higher temperature. Such an abnormal behavior was completely rescued by the expression of human homologue of AHO-3, indicating that the molecular function of AHO-3 is highly conserved between nematode and human. The behavioral regulation by AHO-3 requires the N-terminal cysteine cluster, which ensures the proper subcellular localization of AHO-3 to sensory endings. Double-mutant analysis suggested that AHO-3 acts in the same pathway with ODR-3, a heterotrimeric G protein alpha subunit. Our results unveiled a novel neural protein in C. elegans, confirming its conserved role in behavioral regulation.
Collapse
Affiliation(s)
- Nana Nishio
- Group of Molecular Neurobiology, Division of Biological Science, Graduate School of Science, Nagoya University, Furou-cho, Nagoya 464-8602, Japan
| | | | | | | | | | | | | | | |
Collapse
|
247
|
Myers EM. Gαo and Gαq regulate the expression of daf-7, a TGFβ-like gene, in Caenorhabditis elegans. PLoS One 2012; 7:e40368. [PMID: 22808145 PMCID: PMC3394784 DOI: 10.1371/journal.pone.0040368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/07/2012] [Indexed: 11/20/2022] Open
Abstract
Caenorhabditis elegans enter an alternate developmental stage called dauer in unfavorable conditions such as starvation, overcrowding, or high temperature. Several evolutionarily conserved signaling pathways control dauer formation. DAF-7/TGFβ and serotonin, important ligands in these signaling pathways, affect not only dauer formation, but also the expression of one another. The heterotrimeric G proteins GOA-1 (Gαo) and EGL-30 (Gαq) mediate serotonin signaling as well as serotonin biosynthesis in C. elegans. It is not known whether GOA-1 or EGL-30 also affect dauer formation and/or daf-7 expression, which are both modulated in part by serotonin. The purpose of this study is to better understand the relationship between proteins important for neuronal signaling and developmental plasticity in both C. elegans and humans. Using promoter-GFP transgenic worms, it was determined that both goa-1 and egl-30 regulate daf-7 expression during larval development. In addition, the normal daf-7 response to high temperature or starvation was altered in goa-1 and egl-30 mutants. Despite the effect of goa-1 and egl-30 mutations on daf-7 expression in various environmental conditions, there was no effect of the mutations on dauer formation. This paper provides evidence that while goa-1 and egl-30 are important for normal daf-7 expression, mutations in these genes are not sufficient to disrupt dauer formation.
Collapse
Affiliation(s)
- Edith M Myers
- Department of Biological and Allied Health Sciences, Fairleigh Dickinson University, College at Florham, Madison, New Jersey, United States of America.
| |
Collapse
|
248
|
Cunningham KA, Hua Z, Srinivasan S, Liu J, Lee BH, Edwards RH, Ashrafi K. AMP-activated kinase links serotonergic signaling to glutamate release for regulation of feeding behavior in C. elegans. Cell Metab 2012; 16:113-21. [PMID: 22768843 PMCID: PMC3413480 DOI: 10.1016/j.cmet.2012.05.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 03/08/2012] [Accepted: 05/29/2012] [Indexed: 01/01/2023]
Abstract
Serotonergic regulation of feeding behavior has been studied intensively, both for an understanding of the basic neurocircuitry of energy balance in various organisms and as a therapeutic target for human obesity. However, its underlying molecular mechanisms remain poorly understood. Here, we show that neural serotonin signaling in C. elegans modulates feeding behavior through inhibition of AMP-activated kinase (AMPK) in interneurons expressing the C. elegans counterpart of human SIM1, a transcription factor associated with obesity. In turn, glutamatergic signaling links these interneurons to pharyngeal neurons implicated in feeding behavior. We show that AMPK-mediated regulation of glutamatergic release is conserved in rat hippocampal neurons. These findings reveal cellular and molecular mediators of serotonergic signaling.
Collapse
Affiliation(s)
- Katherine A. Cunningham
- Department of Physiology and Cardiovascular Research Institute and the UCSF Diabetes Center, University of California, San Francisco, San Francisco, California, USA
| | - Zhaolin Hua
- Departments of Physiology and Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Supriya Srinivasan
- Department of Chemical Physiology and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| | - Jason Liu
- Department of Physiology and Cardiovascular Research Institute and the UCSF Diabetes Center, University of California, San Francisco, San Francisco, California, USA
| | - Brian H. Lee
- Department of Physiology and Cardiovascular Research Institute and the UCSF Diabetes Center, University of California, San Francisco, San Francisco, California, USA
| | - Robert H. Edwards
- Departments of Physiology and Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Kaveh Ashrafi
- Department of Physiology and Cardiovascular Research Institute and the UCSF Diabetes Center, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
249
|
Li Y, Yu S, Wu Q, Tang M, Wang D. Transmissions of serotonin, dopamine, and glutamate are required for the formation of neurotoxicity from Al2O3-NPs in nematodeCaenorhabditis elegans. Nanotoxicology 2012; 7:1004-13. [DOI: 10.3109/17435390.2012.689884] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
250
|
Melo JA, Ruvkun G. Inactivation of conserved C. elegans genes engages pathogen- and xenobiotic-associated defenses. Cell 2012; 149:452-66. [PMID: 22500807 DOI: 10.1016/j.cell.2012.02.050] [Citation(s) in RCA: 305] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 01/02/2012] [Accepted: 02/23/2012] [Indexed: 12/18/2022]
Abstract
The nematode C. elegans is attracted to nutritious bacteria and is repelled by pathogens and toxins. Here we show that RNAi and toxin-mediated disruption of core cellular activities, including translation, respiration, and protein turnover, stimulate behavioral avoidance of normally attractive bacteria. RNAi of these and other essential processes induces expression of detoxification and innate immune effectors, even in the absence of toxins or pathogens. Disruption of core processes in non-neuronal tissues was sufficient to stimulate aversion behavior, revealing a neuroendocrine axis of control that additionally required serotonergic and Jnk kinase signaling pathways. We propose that surveillance pathways overseeing core cellular activities allow animals to detect invading pathogens that deploy toxins and virulence factors to undermine vital host functions. Variation in cellular surveillance and endocrine pathways controlling behavior, detoxification, and immunity selected by past toxin or microbial interactions could underlie aberrant responses to foods, medicines, and microbes.
Collapse
Affiliation(s)
- Justine A Melo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|