51
|
Pheromones and Nutritional Signals Regulate the Developmental Reliance on let-7 Family MicroRNAs in C. elegans. Curr Biol 2019; 29:1735-1745.e4. [PMID: 31104929 DOI: 10.1016/j.cub.2019.04.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Adverse environmental conditions can affect rates of animal developmental progression and lead to temporary developmental quiescence (diapause), exemplified by the dauer larva stage of the nematode Caenorhabditis elegans (C. elegans). Remarkably, patterns of cell division and temporal cell-fate progression in C. elegans larvae are not affected by changes in developmental trajectory. However, the underlying physiological and gene regulatory mechanisms that ensure robust developmental patterning despite substantial plasticity in developmental progression are largely unknown. Here, we report that diapause-inducing pheromones correct heterochronic developmental cell lineage defects caused by insufficient expression of let-7 family microRNAs in C. elegans. Moreover, two conserved endocrine signaling pathways, DAF-7/TGF-β and DAF-2/Insulin, that confer on the larva diapause and non-diapause alternative developmental trajectories interact with the nuclear hormone receptor, DAF-12, to initiate and regulate a rewiring of the genetic circuitry controlling temporal cell fates. This rewiring includes engagement of certain heterochronic genes, lin-46, lin-4, and nhl-2, that are previously associated with an altered genetic program in post-diapause animals, in combination with a novel ligand-independent DAF-12 activity, to downregulate the critical let-7 family target Hunchback-like-1 (HBL-1). Our results show how pheromone or endocrine signaling pathways can coordinately regulate both developmental progression and cell-fate transitions in C. elegans larvae under stress so that the developmental schedule of cell fates remains unaffected by changes in developmental trajectory.
Collapse
|
52
|
Butcher RA. Natural products as chemical tools to dissect complex biology in C. elegans. Curr Opin Chem Biol 2019; 50:138-144. [PMID: 31102973 DOI: 10.1016/j.cbpa.2019.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
The search for novel pheromones, hormones, and other types of natural products in the nematode Caenorhabditis elegans has accelerated over the last 10-15 years. Many of these natural products perturb fundamental processes such as developmental progression, metabolism, reproductive and somatic aging, and various behaviors and have thus become essential tools for probing these processes, which are difficult to study in higher organisms. Furthermore, given the similarity between C. elegans and parasitic nematodes, these natural products could potentially be used to manipulate the development and behavior of parasitic nematodes and target the infections caused by them.
Collapse
Affiliation(s)
- Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, FL 32611, United States.
| |
Collapse
|
53
|
Wan X, Zhou Y, Chan CM, Yang H, Yeung C, Chow KL. SRD-1 in AWA neurons is the receptor for female volatile sex pheromones in C. elegans males. EMBO Rep 2019; 20:embr.201846288. [PMID: 30792215 DOI: 10.15252/embr.201846288] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 12/29/2018] [Accepted: 01/22/2019] [Indexed: 01/23/2023] Open
Abstract
Pheromones are critical cues for attracting mating partners for successful reproduction. Sexually mature Caenorhabditis remanei virgin females and self-sperm-depleted Caenorhabditis elegans hermaphrodites produce volatile sex pheromones to attract adult males of both species from afar. The chemoresponsive receptor in males has remained unknown. Here, we show that the male chemotactic behavior requires amphid sensory neurons (AWA neurons) and the G-protein-coupled receptor SRD-1. SRD-1 expression in AWA neurons is sexually dimorphic, with the levels being high in males but undetectable in hermaphrodites. Notably, srd-1 mutant males lack the chemotactic response and pheromone-induced excitation of AWA neurons, both of which can be restored in males and hermaphrodites by AWA-specific srd-1 expression, and ectopic expression of srd-1 in AWB neurons in srd-1 mutants results in a repulsive behavioral response in both sexes. Furthermore, we show that the C-terminal region of SRD-1 confers species-specific differences in the ability to perceive sex pheromones between C. elegans and C. remanei These findings offer an excellent model for dissecting how a single G-protein-coupled receptor expressed in a dimorphic neural system contributes to sex-specific behaviors in animals.
Collapse
Affiliation(s)
- Xuan Wan
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Yuan Zhou
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Chung Man Chan
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Hainan Yang
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Christine Yeung
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - King L Chow
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong .,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong.,Interdisciplinary Programs Office, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| |
Collapse
|
54
|
Park J, Choi W, Dar AR, Butcher RA, Kim K. Neuropeptide Signaling Regulates Pheromone-Mediated Gene Expression of a Chemoreceptor Gene in C. elegans. Mol Cells 2019; 42:28-35. [PMID: 30453729 PMCID: PMC6354054 DOI: 10.14348/molcells.2018.0380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 11/27/2022] Open
Abstract
Animals need to be able to alter their developmental and behavioral programs in response to changing environmental conditions. This developmental and behavioral plasticity is mainly mediated by changes in gene expression. The knowledge of the mechanisms by which environmental signals are transduced and integrated to modulate changes in sensory gene expression is limited. Exposure to ascaroside pheromone has been reported to alter the expression of a subset of putative G protein-coupled chemosensory receptor genes in the ASI chemosensory neurons of C. elegans (Kim et al., 2009; Nolan et al., 2002; Peckol et al., 1999). Here we show that ascaroside pheromone reversibly represses expression of the str-3 chemoreceptor gene in the ASI neurons. Repression of str-3 expression can be initiated only at the L1 stage, but expression is restored upon removal of ascarosides at any developmental stage. Pheromone receptors including SRBC-64/66 and SRG-36/37 are required for str-3 repression. Moreover, pheromone-mediated str-3 repression is mediated by FLP-18 neuropeptide signaling via the NPR-1 neuropeptide receptor. These results suggest that environmental signals regulate chemosensory gene expression together with internal neuropeptide signals which, in turn, modulate behavior.
Collapse
Affiliation(s)
- Jisoo Park
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988,
Korea
| | - Woochan Choi
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988,
Korea
| | - Abdul Rouf Dar
- Department of Chemistry, University of Florida, Gainesville, FL 32611,
USA
| | - Rebecca A. Butcher
- Department of Chemistry, University of Florida, Gainesville, FL 32611,
USA
| | - Kyuhyung Kim
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988,
Korea
| |
Collapse
|
55
|
Shao J, Zhang X, Cheng H, Yue X, Zou W, Kang L. Serotonergic neuron ADF modulates avoidance behaviors by inhibiting sensory neurons in C. elegans. Pflugers Arch 2018; 471:357-363. [PMID: 30206705 DOI: 10.1007/s00424-018-2202-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/29/2018] [Indexed: 10/28/2022]
Abstract
Serotonin plays an essential role in both the invertebrate and vertebrate nervous systems. ADF, an amphid neuron with dual ciliated sensory endings, is considered to be the only serotonergic sensory neuron in the hermaphroditic Caenorhabditis elegans. This neuron is known to be involved in a range of behaviors including pharyngeal pumping, dauer formation, sensory transduction, and memory. However, whether ADF neuron is directly activated by environmental cues and how it processes these information remains unknown. In this study, we found that ADF neuron responds reliably to noxious stimuli such as repulsive odors, copper, sodium dodecyl sulfonate (SDS), and mechanical perturbation. This response is mediated by cell-autonomous and non-cell autonomous mechanisms. Furthermore, we show that ADF can modulate avoidance behaviors by inhibiting ASH, an amphid neuron with single ciliated ending. This work greatly furthers our understanding of 5-HT's contributions to sensory information perception, processing, and the resulting behavioral responses.
Collapse
Affiliation(s)
- Jiajie Shao
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Xiaoyan Zhang
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Hankui Cheng
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Xiaomin Yue
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Wenjuan Zou
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China.
| | - Lijun Kang
- Institute of Neuroscience and Department of Neurosurgery of the First Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Department of Neurobiology, Zhejiang University School of Medicine, 866 Yu Hang Tang Rd., Hangzhou, 310058, Zhejiang, People's Republic of China.
| |
Collapse
|
56
|
McGrath PT, Ruvinsky I. A primer on pheromone signaling in Caenorhabditis elegans for systems biologists. ACTA ACUST UNITED AC 2018; 13:23-30. [PMID: 30984890 DOI: 10.1016/j.coisb.2018.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Individuals communicate information about their age, sex, social status, and recent life history with other members of their species through the release of pheromones, chemical signals that elicit behavioral or physiological changes in the recipients. Pheromones provide a fascinating example of information exchange: animals have evolved intraspecific languages in the presence of eavesdroppers and cheaters. In this review, we discuss the recent work using the nematode C. elegans to decipher its chemical language through the analysis of ascaroside pheromones. Genetic dissection has started to identify the enzymes that produce pheromones and the neural circuits that process these signals. Ecological experiments have characterized the biotic environment of C. elegans and its relatives, including ecological relationships with a variety of species that sense or release similar blends of ascarosides. Systems biology approaches should be fruitful in understanding the organization and function of communication systems in C. elegans.
Collapse
Affiliation(s)
- Patrick T McGrath
- Department of Biological Sciences, Department of Physics; Georgia Institute of Technology, Atlanta, GA 30332.
| | - Ilya Ruvinsky
- Department of Molecular Biosciences; Northwestern University, Evanston, IL 60208.
| |
Collapse
|
57
|
Ma G, Wang T, Korhonen PK, Ang CS, Williamson NA, Young ND, Stroehlein AJ, Hall RS, Koehler AV, Hofmann A, Gasser RB. Molecular alterations during larval development of Haemonchus contortus in vitro are under tight post-transcriptional control. Int J Parasitol 2018; 48:763-772. [PMID: 29792880 DOI: 10.1016/j.ijpara.2018.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/20/2018] [Accepted: 03/26/2018] [Indexed: 12/23/2022]
Abstract
In this study, we explored the molecular alterations in the developmental switch from the L3 to the exsheathed L3 (xL3) and to the L4 stage of Haemonchus contortus in vitro using an integrated transcriptomic, proteomic and bioinformatic approach. Totals of 9,754 mRNAs, 88 microRNAs (miRNAs) and 1,591 proteins were identified, and 6,686 miRNA-mRNA pairs inferred in all larval stages studied. Approximately 16% of transcripts in the combined transcriptome (representing all three larval stages) were expressed as proteins, and there were positive correlations (r = 0.39-0.44) between mRNA transcription and protein expression in the three distinct developmental stages of the parasite. Of the predicted targets, 1,019 (27.0%) mRNA transcripts were expressed as proteins, and there was a negative correlation (r = -0.60 to -0.50) in the differential mRNA transcription and protein expression between developmental stages upon pairwise comparison. The changes in transcription (mRNA and miRNA) and protein expression from the free-living to the parasitic life cycle phase of H. contortus related to enrichments in biological pathways associated with metabolism (e.g., carbohydrate and lipid degradation, and amino acid metabolism), environmental information processing (e.g., signal transduction, signalling molecules and interactions) and/or genetic information processing (e.g., transcription and translation). Specifically, fatty acid degradation, steroid hormone biosynthesis and the Rap1 signalling pathway were suppressed, whereas transcription, translation and protein processing in the endoplasmic reticulum were upregulated during the transition from the free-living L3 to the parasitic xL3 and L4 stages of the nematode in vitro. Dominant post-transcriptional regulation was inferred to elicit these changes, and particular miRNAs (e.g., hco-miR-34 and hco-miR-252) appear to play roles in stress responses and/or environmental adaptations during developmental transitions of H. contortus. Taken together, these integrated results provide a comprehensive insight into the developmental biology of this important parasite at the molecular level in vitro. The approach applied here to H. contortus can be readily applied to other parasitic nematodes.
Collapse
Affiliation(s)
- Guangxu Ma
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nicholas A Williamson
- The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas J Stroehlein
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ross S Hall
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anson V Koehler
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
58
|
Sato K, Sorensen PW. The Chemical Sensitivity and Electrical Activity of Individual Olfactory Sensory Neurons to a Range of Sex Pheromones and Food Odors in the Goldfish. Chem Senses 2018; 43:249-260. [PMID: 29514213 PMCID: PMC5913646 DOI: 10.1093/chemse/bjy016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although it is well established that the olfactory epithelium of teleost fish detects at least 6 classes of biologically relevant odorants using 5 types of olfactory sensory neurons (OSNs), little is understood about the specificity of individual OSNs and thus how they encode identity of natural odors. In this study, we used in vivo extracellular single-unit recording to examine the odor responsiveness and physiological characteristics of 109 individual OSNs in mature male goldfish to a broad range of biological odorants including feeding stimuli (amino acids, polyamines, nucleotides), sex pheromones (sex steroids, prostaglandins [PGs]), and a putative social cue (bile acids). Sixty-one OSNs were chemosensitive, with over half of these (36) responding to amino acids, 7 to polyamines, 7 to nucleotides, 5 to bile acids, 9 to PGs, and 7 to sex steroids. Approximately a quarter of the amino acid-sensitive units also responded to polyamines or nucleotides. Three of 6 amino acid-sensitive units responded to more than 1 amino acid compound, and 5 sex pheromone-sensitive units detected just 1 sex pheromone. While pheromone-sensitive OSNs also responded to the adenylyl cyclase activator, forskolin, amino acid-sensitive OSNs responded to either forskolin or a phospholipase C activator, imipramine. Most OSNs responded to odorants and activators with excitation. Our results suggest that pheromone information is encoded by OSNs specifically tuned to single sex pheromones and employ adenylyl cyclase, suggestive of a labeled-line organization, while food information is encoded by a combination of OSNs that use both adenylyl cyclase and phospholipase C and are often less specifically tuned.
Collapse
Affiliation(s)
- Koji Sato
- Okazaki Institute for Integrative Bioscience, Biosensing Research, Higashiyama Myodaijicho, Okazaki, Aichi, Japan
| | - Peter W Sorensen
- Department of Fisheries, Wildlife and Conservation Biology, University of Minnesota, St Paul, MN, USA
| |
Collapse
|
59
|
Zhang X, Wang Y, Perez DH, Jones Lipinski RA, Butcher RA. Acyl-CoA Oxidases Fine-Tune the Production of Ascaroside Pheromones with Specific Side Chain Lengths. ACS Chem Biol 2018. [PMID: 29537254 DOI: 10.1021/acschembio.7b01021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Caenorhabditis elegans produces a complex mixture of ascaroside pheromones to control its development and behavior. Acyl-CoA oxidases, which participate in β-oxidation cycles that shorten the side chains of the ascarosides, regulate the mixture of pheromones produced. Here, we use CRISPR-Cas9 to make specific nonsense and missense mutations in acox genes and determine the effect of these mutations on ascaroside production in vivo. Ascaroside production in acox-1.1 deletion and nonsense strains, as well as a strain with a missense mutation in a catalytic residue, confirms the central importance of ACOX-1.1 in ascaroside biosynthesis and suggests that ACOX-1.1 functions in part by facilitating the activity of other acyl-CoA oxidases. Ascaroside production in an acox-1.1 strain with a missense mutation in an ATP-binding site at the ACOX-1.1 dimer interface suggests that ATP binding is important for the enzyme to function in ascaroside biosynthesis in vivo. Ascaroside production in strains with deletion, nonsense, and missense mutations in other acox genes demonstrates that ACOX-1.1 works with ACOX-1.3 in processing ascarosides with 7-carbon side chains, ACOX-1.4 in processing ascarosides with 9- and 11-carbon side chains, and ACOX-3 in processing ascarosides with 13- and 15-carbon side chains. It also shows that ACOX-1.2, but not ACOX-1.1, processes ascarosides with 5-carbon ω-side chains. By modeling the ACOX structures, we uncover characteristics of the enzyme active sites that govern substrate preferences. Our work demonstrates the role of specific acyl-CoA oxidases in controlling the length of ascaroside side chains and thus in determining the mixture of pheromones produced by C. elegans.
Collapse
Affiliation(s)
- Xinxing Zhang
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yuting Wang
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - David H. Perez
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | | | - Rebecca A. Butcher
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
60
|
Teuliere J, Kovacevic I, Bao Z, Garriga G. The Caenorhabditis elegans gene ham-1 regulates daughter cell size asymmetry primarily in divisions that produce a small anterior daughter cell. PLoS One 2018; 13:e0195855. [PMID: 29668718 PMCID: PMC5905977 DOI: 10.1371/journal.pone.0195855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/31/2018] [Indexed: 11/30/2022] Open
Abstract
C. elegans cell divisions that produce an apoptotic daughter cell exhibit Daughter Cell Size Asymmetry (DCSA), producing a larger surviving daughter cell and a smaller daughter cell fated to die. Genetic screens for mutants with defects in apoptosis identified several genes that are also required for the ability of these divisions to produce daughter cells that differ in size. One of these genes, ham-1, encodes a putative transcription factor that regulates a subset of the asymmetric cell divisions that produce an apoptotic daughter cell. In a survey of C. elegans divisions, we found that ham-1 mutations affect primarily anterior/posterior divisions that produce a small anterior daughter cell. The affected divisions include those that generate an apoptotic cell as well as those that generate two surviving cells. Our findings suggest that HAM-1 primarily promotes DCSA in a certain class of asymmetric divisions.
Collapse
Affiliation(s)
- Jerome Teuliere
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, United States of America
| | - Ismar Kovacevic
- Developmental Biology Program, Sloan Kettering Institute, New York City, New York, United States of America
| | - Zhirong Bao
- Developmental Biology Program, Sloan Kettering Institute, New York City, New York, United States of America
| | - Gian Garriga
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
61
|
Scheidel N, Kennedy J, Blacque OE. Endosome maturation factors Rabenosyn-5/VPS45 and caveolin-1 regulate ciliary membrane and polycystin-2 homeostasis. EMBO J 2018; 37:embj.201798248. [PMID: 29572244 DOI: 10.15252/embj.201798248] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/08/2018] [Accepted: 02/16/2018] [Indexed: 12/24/2022] Open
Abstract
Primary cilium structure and function relies on control of ciliary membrane homeostasis, regulated by membrane trafficking processes that deliver and retrieve ciliary components at the periciliary membrane. However, the molecular mechanisms controlling ciliary membrane establishment and maintenance, especially in relation to endocytosis, remain poorly understood. Here, using Caenorhabditis elegans, we describe closely linked functions for early endosome (EE) maturation factors RABS-5 (Rabenosyn-5) and VPS-45 (VPS45) in regulating cilium length and morphology, ciliary and periciliary membrane volume, and ciliary signalling-related sensory behaviour. We demonstrate that RABS-5 and VPS-45 control periciliary vesicle number and levels of select EE/endocytic markers (WDFY-2, CAV-1) and the ciliopathy membrane receptor PKD-2 (polycystin-2). Moreover, we show that CAV-1 (caveolin-1) also controls PKD-2 ciliary levels and associated sensory behaviour. These data link RABS-5 and VPS-45 ciliary functions to the processing of periciliary-derived endocytic vesicles and regulation of ciliary membrane homeostasis. Our findings also provide insight into the regulation of PKD-2 ciliary levels via integrated endosomal sorting and CAV-1-mediated endocytosis.
Collapse
Affiliation(s)
- Noémie Scheidel
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Julie Kennedy
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Oliver E Blacque
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
62
|
Fagan KA, Luo J, Lagoy RC, Schroeder FC, Albrecht DR, Portman DS. A Single-Neuron Chemosensory Switch Determines the Valence of a Sexually Dimorphic Sensory Behavior. Curr Biol 2018; 28:902-914.e5. [PMID: 29526590 PMCID: PMC5862148 DOI: 10.1016/j.cub.2018.02.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/11/2018] [Accepted: 02/14/2018] [Indexed: 01/08/2023]
Abstract
Biological sex, a fundamental dimension of internal state, can modulate neural circuits to generate behavioral variation. Understanding how and why circuits are tuned by sex can provide important insights into neural and behavioral plasticity. Here we find that sexually dimorphic behavioral responses to C. elegans ascaroside sex pheromones are implemented by the functional modulation of shared chemosensory circuitry. In particular, the sexual state of a single sensory neuron pair, ADF, determines the nature of an animal's behavioral response regardless of the sex of the rest of the body. Genetic feminization of ADF causes males to be repelled by, rather than attracted to, ascarosides, whereas masculinization of ADF has the opposite effect in hermaphrodites. When ADF is ablated, both sexes are weakly repelled by ascarosides. Genetic sex modulates ADF function by tuning chemosensation: although ADF is functional in both sexes, it detects the ascaroside ascr#3 only in males, a consequence of cell-autonomous action of the master sexual regulator tra-1. This occurs in part through the conserved DM-domain gene mab-3, which promotes the male state of ADF. The sexual modulation of ADF has a key role in reproductive fitness, as feminization or ablation of ADF renders males unable to use ascarosides to locate mates. Our results reveal an economical mechanism in which sex-specific behavioral valence arises through the cell-autonomous regulation of a chemosensory switch by genetic sex, allowing a social cue with salience for both sexes to elicit navigational responses commensurate with the differing needs of each.
Collapse
Affiliation(s)
- Kelli A Fagan
- Neuroscience Graduate Program, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14610, USA
| | - Jintao Luo
- Del Monte Institute for Neuroscience, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14610, USA; Center for Neurotherapeutics Development, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14610, USA
| | - Ross C Lagoy
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Room 4004, Worcester, MA 01605, USA
| | | | - Dirk R Albrecht
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Room 4004, Worcester, MA 01605, USA
| | - Douglas S Portman
- Del Monte Institute for Neuroscience, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14610, USA; Center for Neurotherapeutics Development, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14610, USA; Departments of Biomedical Genetics, Neuroscience, and Biology, University of Rochester, 601 Elmwood Avenue, Box 645, Rochester, NY 14610, USA.
| |
Collapse
|
63
|
O’Donnell MP, Chao PH, Kammenga JE, Sengupta P. Rictor/TORC2 mediates gut-to-brain signaling in the regulation of phenotypic plasticity in C. elegans. PLoS Genet 2018; 14:e1007213. [PMID: 29415022 PMCID: PMC5819832 DOI: 10.1371/journal.pgen.1007213] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/20/2018] [Accepted: 01/22/2018] [Indexed: 01/03/2023] Open
Abstract
Animals integrate external cues with information about internal conditions such as metabolic state to execute the appropriate behavioral and developmental decisions. Information about food quality and quantity is assessed by the intestine and transmitted to modulate neuronal functions via mechanisms that are not fully understood. The conserved Target of Rapamycin complex 2 (TORC2) controls multiple processes in response to cellular stressors and growth factors. Here we show that TORC2 coordinates larval development and adult behaviors in response to environmental cues and feeding state in the bacterivorous nematode C. elegans. During development, pheromone, bacterial food, and temperature regulate expression of the daf-7 TGF-β and daf-28 insulin-like peptide in sensory neurons to promote a binary decision between reproductive growth and entry into the alternate dauer larval stage. We find that TORC2 acts in the intestine to regulate neuronal expression of both daf-7 and daf-28, which together reflect bacterial-diet dependent feeding status, thus providing a mechanism for integration of food signals with external cues in the regulation of neuroendocrine gene expression. In the adult, TORC2 similarly acts in the intestine to modulate food-regulated foraging behaviors via a PDF-2/PDFR-1 neuropeptide signaling-dependent pathway. We also demonstrate that genetic variation affects food-dependent larval and adult phenotypes, and identify quantitative trait loci (QTL) associated with these traits. Together, these results suggest that TORC2 acts as a hub for communication of feeding state information from the gut to the brain, thereby contributing to modulation of neuronal function by internal state. Decision-making in all animals, including humans, involves weighing available information about the external environment as well as the animals’ internal conditions. Information about the environment is obtained via the sensory nervous system, whereas internal state can be assessed via cues such as levels of hormones or nutrients. How multiple external and internal inputs are processed in the nervous system to drive behavior or development is not fully understood. In this study, we examine how the nematode C. elegans integrates dietary information received by the gut with environmental signals to alter nervous system function. We have found that a signaling complex, called TORC2, acts in the gut to relay nutrition signals to alter hormonal signaling by the nervous system in C. elegans. Altered neuronal signaling in turn affects a food-dependent binary developmental decision in larvae, as well as food-dependent foraging behaviors in adults. Our results provide a mechanism by which animals prioritize specific signals such as feeding status to appropriately alter their development and/or behavior.
Collapse
Affiliation(s)
- Michael P. O’Donnell
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA, United States of America
- * E-mail: (MPO); (PS)
| | - Pin-Hao Chao
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA, United States of America
| | - Jan E. Kammenga
- Laboratory of Nematology, Wageningen University and Research, Wageningen, The Netherlands
| | - Piali Sengupta
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA, United States of America
- * E-mail: (MPO); (PS)
| |
Collapse
|
64
|
Vidal B, Aghayeva U, Sun H, Wang C, Glenwinkel L, Bayer EA, Hobert O. An atlas of Caenorhabditis elegans chemoreceptor expression. PLoS Biol 2018; 16:e2004218. [PMID: 29293491 PMCID: PMC5749674 DOI: 10.1371/journal.pbio.2004218] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
One goal of modern day neuroscience is the establishment of molecular maps that assign unique features to individual neuron types. Such maps provide important starting points for neuron classification, for functional analysis, and for developmental studies aimed at defining the molecular mechanisms of neuron identity acquisition and neuron identity diversification. In this resource paper, we describe a nervous system-wide map of the potential expression sites of 244 members of the largest gene family in the C. elegans genome, rhodopsin-like (class A) G-protein-coupled receptor (GPCR) chemoreceptors, using classic gfp reporter gene technology. We cover representatives of all sequence families of chemoreceptor GPCRs, some of which were previously entirely uncharacterized. Most reporters are expressed in a very restricted number of cells, often just in single cells. We assign GPCR reporter expression to all but two of the 37 sensory neuron classes of the sex-shared, core nervous system. Some sensory neurons express a very small number of receptors, while others, particularly nociceptive neurons, coexpress several dozen GPCR reporter genes. GPCR reporters are also expressed in a wide range of inter- and motorneurons, as well as non-neuronal cells, suggesting that GPCRs may constitute receptors not just for environmental signals, but also for internal cues. We observe only one notable, frequent association of coexpression patterns, namely in one nociceptive amphid (ASH) and two nociceptive phasmid sensory neurons (PHA, PHB). We identified GPCRs with sexually dimorphic expression and several GPCR reporters that are expressed in a left/right asymmetric manner. We identified a substantial degree of GPCR expression plasticity; particularly in the context of the environmentally-induced dauer diapause stage when one third of all tested GPCRs alter the cellular specificity of their expression within and outside the nervous system. Intriguingly, in a number of cases, the dauer-specific alterations of GPCR reporter expression in specific neuron classes are maintained during postdauer life and in some case new patterns are induced post-dauer, demonstrating that GPCR gene expression may serve as traits of life history. Taken together, our resource provides an entry point for functional studies and also offers a host of molecular markers for studying molecular patterning and plasticity of the nervous system.
Collapse
Affiliation(s)
- Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Ulkar Aghayeva
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Haosheng Sun
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Chen Wang
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Lori Glenwinkel
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Emily A. Bayer
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
65
|
Early Pheromone Experience Modifies a Synaptic Activity to Influence Adult Pheromone Responses of C. elegans. Curr Biol 2017; 27:3168-3177.e3. [PMID: 28988862 DOI: 10.1016/j.cub.2017.08.068] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/07/2017] [Accepted: 08/29/2017] [Indexed: 11/21/2022]
Abstract
Experiences during early development can influence neuronal functions and modulate adult behaviors [1, 2]. However, the molecular mechanisms underlying the long-term behavioral effects of these early experiences are not fully understood. The C. elegans ascr#3 (asc-ΔC9; C9) pheromone triggers avoidance behavior in adult hermaphrodites [3-7]. Here, we show that hermaphrodites that are briefly exposed to ascr#3 immediately after birth exhibit increased ascr#3-specific avoidance as adults, indicating that ascr#3-experienced animals form a long-lasting memory or imprint of this early ascr#3 exposure [8]. ascr#3 imprinting is mediated by increased synaptic activity between the ascr#3-sensing ADL neurons and their post-synaptic SMB motor neuron partners via increased expression of the odr-2 glycosylated phosphatidylinositol (GPI)-linked signaling gene in the SMB neurons. Our study suggests that the memory for early ascr#3 experience is imprinted via alteration of activity of a single synaptic connection, which in turn shapes experience-dependent plasticity in adult ascr#3 responses.
Collapse
|
66
|
Reilly DK, Lawler DE, Albrecht DR, Srinivasan J. Using an Adapted Microfluidic Olfactory Chip for the Imaging of Neuronal Activity in Response to Pheromones in Male C. Elegans Head Neurons. J Vis Exp 2017. [PMID: 28930991 DOI: 10.3791/56026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The use of calcium indicators has greatly enhanced our understanding of neural dynamics and regulation. The nematode Caenorhabditis elegans, with its completely mapped nervous system and transparent anatomy, presents an ideal model for understanding real-time neural dynamics using calcium indicators. In combination with microfluidic technologies and experimental designs, calcium-imaging studies using these indicators are performed in both free-moving and trapped animals. However, most previous studies utilizing trapping devices, such as the olfactory chip described in Chronis et al., have devices designed for use in the more common hermaphrodite, as the less common male is both morphologically and structurally dissimilar. An adapted olfactory chip was designed and fabricated for increased efficiency in male neuronal imaging with using young adult animals. A turn was incorporated into the worm loading port to rotate the animals and to allow for the separation of the individual neurons within a bilateral pair in 2D imaging. Worms are exposed to a controlled flow of odorant within the microfluidic device, as described in previous hermaphrodite studies. Calcium transients are then analyzed using the open-source software ImageJ. The procedure described herein should allow for an increased amount of male-based C. elegans calcium imaging studies, deepening our understanding of the mechanisms of sex-specific neuronal signaling.
Collapse
Affiliation(s)
- Douglas K Reilly
- Department of Biology and Biotechnology, Worcester Polytechnic Institute
| | - Daniel E Lawler
- Department of Biomedical Engineering, Worcester Polytechnic Institute
| | - Dirk R Albrecht
- Department of Biology and Biotechnology, Worcester Polytechnic Institute; Department of Biomedical Engineering, Worcester Polytechnic Institute;
| | - Jagan Srinivasan
- Department of Biology and Biotechnology, Worcester Polytechnic Institute
| |
Collapse
|
67
|
Aprison EZ, Ruvinsky I. Counteracting Ascarosides Act through Distinct Neurons to Determine the Sexual Identity of C. elegans Pheromones. Curr Biol 2017; 27:2589-2599.e3. [DOI: 10.1016/j.cub.2017.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/26/2017] [Accepted: 07/13/2017] [Indexed: 01/12/2023]
|
68
|
Park S, Paik YK. Genetic deficiency in neuronal peroxisomal fatty acid β-oxidation causes the interruption of dauer development in Caenorhabditis elegans. Sci Rep 2017; 7:9358. [PMID: 28839231 PMCID: PMC5571181 DOI: 10.1038/s41598-017-10020-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/02/2017] [Indexed: 11/11/2022] Open
Abstract
Although peroxisomal fatty acid (FA) β-oxidation is known to be critical for animal development, the cellular mechanisms that control the manner in which its neuronal deficiency causes developmental defects remain unclear. To elucidate the potential cellular consequences of neuronal FA metabolic disorder for dauer development, an alternative developmental process in Caenorhabditis elegans that occurs during stress, we investigated the sequential effects of its corresponding genetic deficiency. Here, we show that the daf-22 gene in peroxisomal FA β-oxidation plays a distinct role in ASK neurons, and its deficiency interrupts dauer development even in the presence of the exogenous ascaroside pheromones that induce such development. Un-metabolized FAs accumulated in ASK neurons of daf-22 mutants stimulate the endoplasmic reticulum (ER) stress response, which may enhance the XBP-1 activity that promotes the transcription of neuronal insulin-like peptides. These sequential cell-autonomous reactions in ASK neurons then activate insulin/IGF-1 signaling, which culminates in the suppression of DAF-16/FOXO activity. This suppression results in the interruption of dauer development, independently of pheromone presence. These findings suggest that neuronal peroxisomal FA β-oxidation is indispensable for animal development by regulating the ER stress response and neuroendocrine signaling.
Collapse
Affiliation(s)
- Saeram Park
- Department of Integrated OMICS for Biomedical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young-Ki Paik
- Department of Integrated OMICS for Biomedical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Department of Biochemistry, College of Life Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
69
|
A conserved neuronal DAF-16/FoxO plays an important role in conveying pheromone signals to elicit repulsion behavior in Caenorhabditis elegans. Sci Rep 2017; 7:7260. [PMID: 28775361 PMCID: PMC5543152 DOI: 10.1038/s41598-017-07313-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 01/09/2023] Open
Abstract
Animals use pheromones as a conspecific chemical language to respond appropriately to environmental changes. The soil nematode Caenorhabditis elegans secretes ascaroside pheromones throughout the lifecycle, which influences entry into dauer phase in early larvae, in addition to sexual attraction and aggregation. In adult hermaphrodites, pheromone sensory signals perceived by worms usually elicit repulsion as an initial behavioral signature. However, the molecular mechanisms underlying neuronal pheromone sensory process from perception to repulsion in adult hermaphrodites remain poorly understood. Here, we show that pheromone signals perceived by GPA-3 is conveyed through glutamatergic neurotransmission in which neuronal DAF-16/FoxO plays an important modulatory role by controlling glutaminase gene expression. We further provide evidence that this modulatory role for DAF-16/FoxO seems to be conserved evolutionarily by electro-physiological study in mouse primary hippocampal neurons that are responsible for glutamatergic neurotransmission. These findings provide the basis for understanding the nematode pheromone signaling, which seems crucial for adaptation of adult hermaphrodites to changes in environmental condition for survival.
Collapse
|
70
|
Small-molecule pheromones and hormones controlling nematode development. Nat Chem Biol 2017; 13:577-586. [PMID: 28514418 DOI: 10.1038/nchembio.2356] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/15/2017] [Indexed: 12/21/2022]
Abstract
The existence of small-molecule signals that influence development in Caenorhabditis elegans has been known for several decades, but only in recent years have the chemical structures of several of these signals been established. The identification of these signals has enabled connections to be made between these small molecules and fundamental signaling pathways in C. elegans that influence not only development but also metabolism, fertility, and lifespan. Spurred by these important discoveries and aided by recent advances in comparative metabolomics and NMR spectroscopy, the field of nematode chemistry has the potential to expand dramatically in the coming years. This Perspective will focus on small-molecule pheromones and hormones that influence developmental events in the nematode life cycle (ascarosides, dafachronic acids, and nemamides), will cover more recent work regarding the biosynthesis of these signals, and will explore how the discovery of these signals is transforming our understanding of nematode development and physiology.
Collapse
|
71
|
Hussey R, Stieglitz J, Mesgarzadeh J, Locke TT, Zhang YK, Schroeder FC, Srinivasan S. Pheromone-sensing neurons regulate peripheral lipid metabolism in Caenorhabditis elegans. PLoS Genet 2017; 13:e1006806. [PMID: 28545126 PMCID: PMC5456406 DOI: 10.1371/journal.pgen.1006806] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 06/02/2017] [Accepted: 05/05/2017] [Indexed: 12/03/2022] Open
Abstract
It is now established that the central nervous system plays an important role in regulating whole body metabolism and energy balance. However, the extent to which sensory systems relay environmental information to modulate metabolic events in peripheral tissues has remained poorly understood. In addition, it has been challenging to map the molecular mechanisms underlying discrete sensory modalities with respect to their role in lipid metabolism. In previous work our lab has identified instructive roles for serotonin signaling as a surrogate for food availability, as well as oxygen sensing, in the control of whole body metabolism. In this study, we now identify a role for a pair of pheromone-sensing neurons in regulating fat metabolism in C. elegans, which has emerged as a tractable and highly informative model to study the neurobiology of metabolism. A genetic screen revealed that GPA-3, a member of the Gα family of G proteins, regulates body fat content in the intestine, the major metabolic organ for C. elegans. Genetic and reconstitution studies revealed that the potent body fat phenotype of gpa-3 null mutants is controlled from a pair of neurons called ADL(L/R). We show that cAMP functions as the second messenger in the ADL neurons, and regulates body fat stores via the neurotransmitter acetylcholine, from downstream neurons. We find that the pheromone ascr#3, which is detected by the ADL neurons, regulates body fat stores in a GPA-3-dependent manner. We define here a third sensory modality, pheromone sensing, as a major regulator of body fat metabolism. The pheromone ascr#3 is an indicator of population density, thus we hypothesize that pheromone sensing provides a salient 'denominator' to evaluate the amount of food available within a population and to accordingly adjust metabolic rate and body fat levels. The central nervous system plays a vital role in regulating whole body metabolism and energy balance. However, the precise cellular, genetic and molecular mechanisms underlying these effects remain a major unsolved mystery. C. elegans has emerged as a tractable and highly informative model to study the neurobiology of metabolism. Previously, we have identified instructive roles for serotonin signaling as a surrogate for food availability, as well as oxygen sensing, in the control of whole body metabolism. In our current study we have identified a role for a pair of pheromone-sensing neurons in regulating fat metabolism in C. elegans. cAMP acts as a second messenger in these neurons, and regulates body fat stores via acetylcholine signaling in the nervous system. We find that the population-density-sensing pheromone detected by these neurons regulates body fat stores. Together, we define a third sensory modality, population density sensing, as a major regulator of body fat metabolism.
Collapse
Affiliation(s)
- Rosalind Hussey
- Department of Molecular Medicine and Department of Neuroscience, The Scripps Research Institute, La Jolla, California, United States of America
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jon Stieglitz
- Kellogg School of Science and Technology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jaleh Mesgarzadeh
- Department of Molecular Medicine and Department of Neuroscience, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Tiffany T. Locke
- Department of Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Ying K. Zhang
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Frank C. Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Supriya Srinivasan
- Department of Molecular Medicine and Department of Neuroscience, The Scripps Research Institute, La Jolla, California, United States of America
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
72
|
Abstract
The nematode Caenorhabditis elegans produces tens, if not hundreds, of different ascarosides as pheromones to communicate with other members of its species. Overlapping mixtures of these pheromones affect the development of the worm and a variety of different behaviors. The ascarosides represent a unique tool for dissecting the neural circuitry that controls behavior and that connects to important signaling pathways, such as the insulin and TGFβ pathways, that lie at the nexus of development, metabolism, and lifespan in C. elegans. However, the exact physiological roles of many of the ascarosides are unclear, especially since many of these pheromones likely have multiple functions depending on their concentrations, the presence of other pheromones, and a variety of other factors. Determining these physiological roles will be facilitated by top-down approaches to characterize the pheromone receptors and their function, as well as bottom-up approaches to characterize the pheromone biosynthetic enzymes and their regulation.
Collapse
Affiliation(s)
- Rebecca A Butcher
- University of Florida, Department of Chemistry, Gainesville, FL 32611, USA.
| |
Collapse
|
73
|
Cheng L, Xu S, Xu C, Lu H, Zhang Z, Zhang D, Mu W, Liu F. Effects of trans-2-hexenal on reproduction, growth and behaviour and efficacy against the pinewood nematode, Bursaphelenchus xylophilus. PEST MANAGEMENT SCIENCE 2017; 73:888-895. [PMID: 27414986 DOI: 10.1002/ps.4360] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/12/2016] [Accepted: 07/12/2016] [Indexed: 05/02/2023]
Abstract
BACKGROUND Bursaphelenchus xylophilus is a serious quarantined pest that causes severe damage and major economic losses to pine forests. Because of the adverse effects of some traditional nematicides on humans and the environment, the search for new plant toxicants against these nematodes has intensified. Nematicidal activity of trans-2-hexenal, which is a six-carbon aldehyde present in many plants, was tested against the nematode. RESULTS trans-2-Hexenal showed significant efficacy against B. xylophilus in a dose range of 349.5-699 g m-3 by fumigation of pinewood logs. Additionally, it had significant nematicidal activity against different life stages of B. xylophilus in an in vitro test, with second-stage larvae (L2s) being the most sensitive, with an LC50 value of 9.87 µg mL-1 at 48 h. Egg hatch was also significantly inhibited. Further studies revealed that trans-2-hexenal inhibited the reproductive activity of B. xylophilus, with negative effects on reproduction rate and egg numbers. Moreover, trans-2-hexenal reduced the body length of B. xylophilus. Respiratory rate and thrashing behaviour of B. xylophilus also decreased following treatment with this compound. CONCLUSION trans-2-Hexenal had significant nematicidal activity against B. xylophilus, providing a basis for elucidation of the mode of action of trans-2-hexenal against plant-parasitic nematodes in future studies. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Le Cheng
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Shuangyu Xu
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Chunmei Xu
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Hongbao Lu
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Zhengqun Zhang
- College of Horticultural Science and Engineering, Shandong Agricultural University, Tai'an, Shandong, China
| | - Daxia Zhang
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Wei Mu
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| | - Feng Liu
- Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Tai'an, Shandong, China
| |
Collapse
|
74
|
Bharadwaj PS, Hall SE. Endogenous RNAi Pathways Are Required in Neurons for Dauer Formation in Caenorhabditis elegans. Genetics 2017; 205:1503-1516. [PMID: 28122825 PMCID: PMC5378109 DOI: 10.1534/genetics.116.195438] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/21/2017] [Indexed: 12/16/2022] Open
Abstract
Animals can adapt to unfavorable environments through changes in physiology or behavior. In the nematode, Caenorhabditis elegans, environmental conditions perceived early in development determine whether the animal enters either the reproductive cycle, or enters into an alternative diapause stage named dauer. Here, we show that endogenous RNAi pathways play a role in dauer formation in crowding (high pheromone), starvation, and high temperature conditions. Disruption of the Mutator proteins or the nuclear Argonaute CSR-1 result in differential dauer-deficient phenotypes that are dependent upon the experienced environmental stress. We provide evidence that the RNAi pathways function in chemosensory neurons for dauer formation, upstream of the TGF-β and insulin signaling pathways. In addition, we show that Mutator MUT-16 expression in a subset of individual pheromone-sensing neurons is sufficient for dauer formation in high pheromone conditions, but not in starvation or high temperature conditions. Furthermore, we also show that MUT-16 and CSR-1 are required for expression of a subset of G proteins with functions in the detection of pheromone components. Together, our data suggest a model where Mutator-amplified siRNAs that associate with the CSR-1 pathway promote expression of genes required for the detection and signaling of environmental conditions to regulate development and behavior in C. elegans This study highlights a mechanism whereby RNAi pathways mediate the link between environmental stress and adaptive phenotypic plasticity in animals.
Collapse
Affiliation(s)
| | - Sarah E Hall
- Department of Biology, Syracuse University, New York 13244
| |
Collapse
|
75
|
Ghosh DD, Nitabach MN, Zhang Y, Harris G. Multisensory integration in C. elegans. Curr Opin Neurobiol 2017; 43:110-118. [PMID: 28273525 PMCID: PMC5501174 DOI: 10.1016/j.conb.2017.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/19/2022]
Abstract
Multisensory integration is a neural process by which signals from two or more distinct sensory channels are simultaneously processed to form a more coherent representation of the environment. Multisensory integration, especially when combined with a survey of internal states, provides selective advantages for animals navigating complex environments. Despite appreciation of the importance of multisensory integration in behavior, the underlying molecular and cellular mechanisms remain poorly understood. Recent work looking at how Caenorhabditis elegans makes multisensory decisions has yielded mechanistic insights into how a relatively simple and well-defined nervous system employs circuit motifs of defined features, synaptic signals and extrasynaptic neurotransmission, as well as neuromodulators in processing and integrating multiple sensory inputs to generate flexible and adaptive behavioral outputs.
Collapse
Affiliation(s)
- D Dipon Ghosh
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
| | - Michael N Nitabach
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, United States; Department of Genetics, Yale University, New Haven, CT, United States; Kavli Institute for Neuroscience, Yale University, New Haven, CT, United States.
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States.
| | - Gareth Harris
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States
| |
Collapse
|
76
|
Molecular Determinants of the Regulation of Development and Metabolism by Neuronal eIF2α Phosphorylation in Caenorhabditis elegans. Genetics 2017; 206:251-263. [PMID: 28292919 DOI: 10.1534/genetics.117.200568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/09/2017] [Indexed: 11/18/2022] Open
Abstract
Cell-nonautonomous effects of signaling in the nervous system of animals can influence diverse aspects of organismal physiology. We previously showed that phosphorylation of Ser49 of the α-subunit of eukaryotic translation initiation factor 2 (eIF2α) in two chemosensory neurons by PEK-1/PERK promotes entry of Caenorhabditis elegans into dauer diapause. Here, we identified and characterized the molecular determinants that confer sensitivity to effects of neuronal eIF2α phosphorylation on development and physiology of C. elegans Isolation and characterization of mutations in eif-2Ba encoding the α-subunit of eIF2B support a conserved role, previously established by studies in yeast, for eIF2Bα in providing a binding site for phosphorylated eIF2α to inhibit the exchange factor eIF2B catalytic activity that is required for translation initiation. We also identified a mutation in eif-2c, encoding the γ-subunit of eIF2, which confers insensitivity to the effects of phosphorylated eIF2α while also altering the requirement for eIF2Bγ. In addition, we show that constitutive expression of eIF2α carrying a phosphomimetic S49D mutation in the ASI pair of sensory neurons confers dramatic effects on growth, metabolism, and reproduction in adult transgenic animals, phenocopying systemic responses to starvation. Furthermore, we show that constitutive expression of eIF2α carrying a phosphomimetic S49D mutation in the ASI neurons enhances dauer entry through bypassing the requirement for nutritionally deficient conditions. Our data suggest that the state of eIF2α phosphorylation in the ASI sensory neuron pair may modulate internal nutrient sensing and signaling pathways, with corresponding organismal effects on development and metabolism.
Collapse
|
77
|
Palamiuc L, Noble T, Witham E, Ratanpal H, Vaughan M, Srinivasan S. A tachykinin-like neuroendocrine signalling axis couples central serotonin action and nutrient sensing with peripheral lipid metabolism. Nat Commun 2017; 8:14237. [PMID: 28128367 PMCID: PMC5290170 DOI: 10.1038/ncomms14237] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 12/09/2016] [Indexed: 01/13/2023] Open
Abstract
Serotonin, a central neuromodulator with ancient ties to feeding and metabolism, is a major driver of body fat loss. However, mechanisms by which central serotonin action leads to fat loss remain unknown. Here, we report that the FLP-7 neuropeptide and its cognate receptor, NPR-22, function as the ligand-receptor pair that defines the neuroendocrine axis of serotonergic body fat loss in Caenorhabditis elegans. FLP-7 is secreted as a neuroendocrine peptide in proportion to fluctuations in neural serotonin circuit functions, and its release is regulated from secretory neurons via the nutrient sensor AMPK. FLP-7 acts via the NPR-22/Tachykinin2 receptor in the intestine and drives fat loss via the adipocyte triglyceride lipase ATGL-1. Importantly, this ligand-receptor pair does not alter other serotonin-dependent behaviours including food intake. For global modulators such as serotonin, the use of distinct neuroendocrine peptides for each output may be one means to achieve phenotypic selectivity.
Collapse
Affiliation(s)
- Lavinia Palamiuc
- Department of Chemical Physiology and The Dorris Neuroscience Center, 1 Barnard Drive, Oceanside, California 92056, USA
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Tallie Noble
- Mira Costa College, 1 Barnard Drive, Oceanside, California 92056, USA
| | - Emily Witham
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Harkaranveer Ratanpal
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Megan Vaughan
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
- Kellogg School of Science and Technology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Supriya Srinivasan
- Department of Chemical Physiology and The Dorris Neuroscience Center, 1 Barnard Drive, Oceanside, California 92056, USA
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| |
Collapse
|
78
|
Hsueh YP, Gronquist MR, Schwarz EM, Nath RD, Lee CH, Gharib S, Schroeder FC, Sternberg PW. Nematophagous fungus Arthrobotrys oligospora mimics olfactory cues of sex and food to lure its nematode prey. eLife 2017; 6. [PMID: 28098555 PMCID: PMC5243009 DOI: 10.7554/elife.20023] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 12/28/2016] [Indexed: 12/19/2022] Open
Abstract
To study the molecular basis for predator-prey coevolution, we investigated how Caenorhabditis elegans responds to the predatory fungus Arthrobotrys oligospora. C. elegans and other nematodes were attracted to volatile compounds produced by A. oligospora. Gas-chromatographic mass-spectral analyses of A. oligospora-derived volatile metabolites identified several odors mimicking food cues attractive to nematodes. One compound, methyl 3-methyl-2-butenoate (MMB) additionally triggered strong sex- and stage-specific attraction in several Caenorhabditis species. Furthermore, when MMB is present, it interferes with nematode mating, suggesting that MMB might mimic sex pheromone in Caenorhabditis species. Forward genetic screening suggests that multiple receptors are involved in sensing MMB. Response to fungal odors involves the olfactory neuron AWCs. Single-cell RNA-seq revealed the GPCRs expressed in AWC. We propose that A. oligospora likely evolved the means to use olfactory mimicry to attract its nematode prey through the olfactory neurons in C. elegans and related species. DOI:http://dx.doi.org/10.7554/eLife.20023.001
Collapse
Affiliation(s)
- Yen-Ping Hsueh
- Division of Biology and Biological Engineering, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Matthew R Gronquist
- Department of Chemistry, State University of New York at Fredonia, Fredonia, United States
| | - Erich M Schwarz
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Ravi David Nath
- Division of Biology and Biological Engineering, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States
| | - Ching-Han Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Shalha Gharib
- Division of Biology and Biological Engineering, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, United States.,Department of Chemistry and Chemical Biology, Cornell University, Ithaca, United States
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States
| |
Collapse
|
79
|
Lok JB. Signaling in Parasitic Nematodes: Physicochemical Communication Between Host and Parasite and Endogenous Molecular Transduction Pathways Governing Worm Development and Survival. CURRENT CLINICAL MICROBIOLOGY REPORTS 2016; 3:186-197. [PMID: 28781934 PMCID: PMC5543980 DOI: 10.1007/s40588-016-0046-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Signaling or communication between host and parasite may occur over relatively long ranges to enable host finding and acquisition by infective parasitic nematode larvae. Innate behaviors in infective larvae transmitted from the soil that enhance the likelihood of host contact, such as negative geotaxis and hypermotility, are likely mediated by mechanoreception and neuromuscular signaling. Host cues such as vibration of the substratum, elevated temperature, exhaled CO2, and other volatile odorants are perceived by mechanosensory and chemosensory neurons of the amphidial complex. Beyond this, the molecular systems that transduce these external cues within the worm are unknown at this time. Overall, the signal transduction mechanisms that regulate switching between dauer and continuous reproductive development in Caenorhabditis elegans, and doubtless other free-living nematodes, have provided a useful framework for testing hypotheses about how the morphogenesis and development of infective parasitic nematode larvae and the lifespan of adult parasites are regulated. In C. elegans, four major signal transduction pathways, G protein-coupled receptor signaling, insulin/insulin-like growth factor signaling, TGFβ-like signaling and steroid-nuclear hormone receptor signaling govern the switch between dauer and continuous development and regulate adult lifespan. Parasitic nematodes appear to have conserved the functions of G-protein-coupled signaling, insulin-like signaling and steroid-nuclear hormone receptor signaling to regulate larval development before and during the infective process. By contrast, TGFβ-like signaling appears to have been adapted for some other function, perhaps modulation of the host immune response. Of the three signal transduction pathways that appear to regulate development in parasitic nematodes, steroid-nuclear hormone signaling is the most straightforward to manipulate with administered small molecules and may form the basis of new chemotherapeutic strategies. Signaling between parasites and their hosts' immune systems also occurs and serves to modulate these responses to allow chronic infection and down regulate acute inflammatory responses. Knowledge of the precise nature of this signaling may form the basis of immunological interventions to protect against parasitism or related lesions and to alleviate inflammatory diseases of various etiologies.
Collapse
Affiliation(s)
- James B Lok
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104 USA
| |
Collapse
|
80
|
Greene JS, Dobosiewicz M, Butcher RA, McGrath PT, Bargmann CI. Regulatory changes in two chemoreceptor genes contribute to a Caenorhabditis elegans QTL for foraging behavior. eLife 2016; 5. [PMID: 27893361 PMCID: PMC5125752 DOI: 10.7554/elife.21454] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/14/2016] [Indexed: 12/22/2022] Open
Abstract
Natural isolates of C. elegans differ in their sensitivity to pheromones that inhibit exploratory behavior. Previous studies identified a QTL for pheromone sensitivity that includes alternative alleles of srx-43, a chemoreceptor that inhibits exploration through its activity in ASI sensory neurons. Here we show that the QTL is multigenic and includes alternative alleles of srx-44, a second chemoreceptor gene that modifies pheromone sensitivity. srx-44 either promotes or inhibits exploration depending on its expression in the ASJ or ADL sensory neurons, respectively. Naturally occurring pheromone insensitivity results in part from previously described changes in srx-43 expression levels, and in part from increased srx-44 expression in ASJ, which antagonizes ASI and ADL. Antagonism between the sensory neurons results in cellular epistasis that is reflected in their transcription of insulin genes that regulate exploration. These results and genome-wide evidence suggest that chemoreceptor genes may be preferred sites of adaptive variation in C. elegans. DOI:http://dx.doi.org/10.7554/eLife.21454.001
Collapse
Affiliation(s)
- Joshua S Greene
- Howard Hughes Medical Institute (HHMI), Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, United States
| | - May Dobosiewicz
- Howard Hughes Medical Institute (HHMI), Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, United States
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Patrick T McGrath
- Department of Biology, Georgia Institute of Technology, Atlanta, United States
| | - Cornelia I Bargmann
- Howard Hughes Medical Institute (HHMI), Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, United States
| |
Collapse
|
81
|
Greene JS, Brown M, Dobosiewicz M, Ishida IG, Macosko EZ, Zhang X, Butcher RA, Cline DJ, McGrath PT, Bargmann CI. Balancing selection shapes density-dependent foraging behaviour. Nature 2016; 539:254-258. [PMID: 27799655 PMCID: PMC5161598 DOI: 10.1038/nature19848] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 09/14/2016] [Indexed: 02/04/2023]
Abstract
The optimal foraging strategy in a given environment depends on the number of competing individuals and their behavioural strategies. Little is known about the genes and neural circuits that integrate social information into foraging decisions. Here we show that ascaroside pheromones, small glycolipids that signal population density, suppress exploratory foraging in Caenorhabditis elegans, and that heritable variation in this behaviour generates alternative foraging strategies. We find that natural C. elegans isolates differ in their sensitivity to the potent ascaroside icas#9 (IC-asc-C5). A quantitative trait locus (QTL) regulating icas#9 sensitivity includes srx-43, a G-protein-coupled icas#9 receptor that acts in the ASI class of sensory neurons to suppress exploration. Two ancient haplotypes associated with this QTL confer competitive growth advantages that depend on ascaroside secretion, its detection by srx-43 and the distribution of food. These results suggest that balancing selection at the srx-43 locus generates alternative density-dependent behaviours, fulfilling a prediction of foraging game theory.
Collapse
Affiliation(s)
- Joshua S Greene
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - Maximillian Brown
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - May Dobosiewicz
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - Itzel G Ishida
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - Evan Z Macosko
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - Xinxing Zhang
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, USA
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, USA
| | - Devin J Cline
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Patrick T McGrath
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Cornelia I Bargmann
- Howard Hughes Medical Institute, Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
82
|
Large EE, Xu W, Zhao Y, Brady SC, Long L, Butcher RA, Andersen EC, McGrath PT. Selection on a Subunit of the NURF Chromatin Remodeler Modifies Life History Traits in a Domesticated Strain of Caenorhabditis elegans. PLoS Genet 2016; 12:e1006219. [PMID: 27467070 PMCID: PMC4965130 DOI: 10.1371/journal.pgen.1006219] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/01/2016] [Indexed: 11/20/2022] Open
Abstract
Evolutionary life history theory seeks to explain how reproductive and survival traits are shaped by selection through allocations of an individual’s resources to competing life functions. Although life-history traits evolve rapidly, little is known about the genetic and cellular mechanisms that control and couple these tradeoffs. Here, we find that two laboratory-adapted strains of C. elegans descended from a single common ancestor that lived in the 1950s have differences in a number of life-history traits, including reproductive timing, lifespan, dauer formation, growth rate, and offspring number. We identified a quantitative trait locus (QTL) of large effect that controls 24%–75% of the total trait variance in reproductive timing at various timepoints. Using CRISPR/Cas9-induced genome editing, we show this QTL is due in part to a 60 bp deletion in the 3’ end of the nurf-1 gene, which is orthologous to the human gene encoding the BPTF component of the NURF chromatin remodeling complex. Besides reproduction, nurf-1 also regulates growth rate, lifespan, and dauer formation. The fitness consequences of this deletion are environment specific—it increases fitness in the growth conditions where it was fixed but decreases fitness in alternative laboratory growth conditions. We propose that chromatin remodeling, acting through nurf-1, is a pleiotropic regulator of life history trade-offs underlying the evolution of multiple traits across different species. Sex and death are two fundamental concerns of each organism. These traits evolve rapidly in natural populations as animals seek to maximize their fitness in a given environment. For example, in mammals, lifespan, size, and fecundity vary over two order of magnitude. A key observation of evolutionary life history theory is the recognition that there are limited amount of resources available, which creates tradeoffs between competing life functions. By studying a domesticated strain of C. elegans, we identify a beneficial mutation that regulates a number of life history tradeoffs. This mutation affects a subunit of the NURF chromatin remodeling complex. Our work suggests that NURF is a master regulator of life history tradeoffs through epigenetic regulation, and a target of evolution.
Collapse
Affiliation(s)
- Edward E. Large
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Wen Xu
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Yuehui Zhao
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Shannon C. Brady
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Lijiang Long
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Rebecca A. Butcher
- Department of Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Erik C. Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Patrick T. McGrath
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
83
|
Krzyzanowski MC, Woldemariam S, Wood JF, Chaubey AH, Brueggemann C, Bowitch A, Bethke M, L’Etoile ND, Ferkey DM. Aversive Behavior in the Nematode C. elegans Is Modulated by cGMP and a Neuronal Gap Junction Network. PLoS Genet 2016; 12:e1006153. [PMID: 27459302 PMCID: PMC4961389 DOI: 10.1371/journal.pgen.1006153] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/08/2016] [Indexed: 01/03/2023] Open
Abstract
All animals rely on their ability to sense and respond to their environment to survive. However, the suitability of a behavioral response is context-dependent, and must reflect both an animal's life history and its present internal state. Based on the integration of these variables, an animal's needs can be prioritized to optimize survival strategies. Nociceptive sensory systems detect harmful stimuli and allow for the initiation of protective behavioral responses. The polymodal ASH sensory neurons are the primary nociceptors in C. elegans. We show here that the guanylyl cyclase ODR-1 functions non-cell-autonomously to downregulate ASH-mediated aversive behaviors and that ectopic cGMP generation in ASH is sufficient to dampen ASH sensitivity. We define a gap junction neural network that regulates nociception and propose that decentralized regulation of ASH signaling can allow for rapid correlation between an animal's internal state and its behavioral output, lending modulatory flexibility to this hard-wired nociceptive neural circuit.
Collapse
Affiliation(s)
- Michelle C. Krzyzanowski
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Sarah Woldemariam
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Jordan F. Wood
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Aditi H. Chaubey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Chantal Brueggemann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Alexander Bowitch
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Mary Bethke
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Noelle D. L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Denise M. Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| |
Collapse
|
84
|
Reis Rodrigues P, Kaul TK, Ho JH, Lucanic M, Burkewitz K, Mair WB, Held JM, Bohn LM, Gill MS. Synthetic Ligands of Cannabinoid Receptors Affect Dauer Formation in the Nematode Caenorhabditis elegans. G3 (BETHESDA, MD.) 2016; 6:1695-705. [PMID: 27172180 PMCID: PMC4889665 DOI: 10.1534/g3.116.026997] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/04/2016] [Indexed: 01/20/2023]
Abstract
Under adverse environmental conditions the nematode Caenorhabditis elegans can enter an alternate developmental stage called the dauer larva. To identify lipophilic signaling molecules that influence this process, we screened a library of bioactive lipids and found that AM251, an antagonist of the human cannabinoid (CB) receptor, suppresses dauer entry in daf-2 insulin receptor mutants. AM251 acted synergistically with glucose supplementation indicating that the metabolic status of the animal influenced the activity of this compound. Similarly, loss of function mutations in the energy-sensing AMP-activated kinase subunit, aak-2, enhanced the dauer-suppressing effects of AM251, while constitutive activation of aak-2 in neurons was sufficient to inhibit AM251 activity. Chemical epistasis experiments indicated that AM251 acts via G-protein signaling and requires the TGF-β ligand DAF-7, the insulin peptides DAF-28 and INS-6, and a functional ASI neuron to promote reproductive growth. AM251 also required the presence of the SER-5 serotonin receptor, but in vitro experiments suggest that this may not be via a direct interaction. Interestingly, we found that other antagonists of mammalian CB receptors also suppress dauer entry, while the nonselective CB receptor agonist, O-2545, not only inhibited the activity of AM251, but also was able to promote dauer entry when administered alone. Since worms do not have obvious orthologs of CB receptors, the effects of synthetic CBs on neuroendocrine signaling in C. elegans are likely to be mediated via another, as yet unknown, receptor mechanism. However, we cannot exclude the existence of a noncanonical CB receptor in C. elegans.
Collapse
Affiliation(s)
- Pedro Reis Rodrigues
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida 33458
| | - Tiffany K Kaul
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida 33458
| | - Jo-Hao Ho
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458
| | - Mark Lucanic
- The Buck Institute for Research on Aging, Novato, California 94945
| | - Kristopher Burkewitz
- Department of Genetics and Complex Diseases, School of Public Health, Harvard University, Boston, Massachusetts 02115
| | - William B Mair
- Department of Genetics and Complex Diseases, School of Public Health, Harvard University, Boston, Massachusetts 02115
| | - Jason M Held
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110 Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Laura M Bohn
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458
| | - Matthew S Gill
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida 33458
| |
Collapse
|
85
|
Leighton DH, Sternberg PW. Mating pheromones of Nematoda: olfactory signaling with physiological consequences. Curr Opin Neurobiol 2016; 38:119-24. [PMID: 27213246 DOI: 10.1016/j.conb.2016.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 01/17/2023]
Abstract
Secreted pheromones have long been known to influence mating in the phylum Nematoda. The study of nematode sexual behavior has greatly benefited in the last decade from the genetic and neurobiological tools available for the model nematode Caenorhabditis elegans, as well as from the chemical identification of many pheromones secreted by this species. The discovery that nematodes can influence one another's physiological development and stress responsiveness through the sharing of pheromones, in addition to simply triggering sexual attraction, is particularly striking. Here we review recent research on nematode mating pheromones, which has been conducted predominantly on C. elegans, but there are beginning to be parallel studies in other species.
Collapse
Affiliation(s)
- Daniel Hw Leighton
- HHMI and Division of Biology and Biological Engineering, Caltech, Pasadena 91125, USA
| | - Paul W Sternberg
- HHMI and Division of Biology and Biological Engineering, Caltech, Pasadena 91125, USA.
| |
Collapse
|
86
|
Abstract
DNA does not make phenotypes on its own. In this volume entitled "Genes and Phenotypic Evolution," the present review draws the attention on the process of phenotype construction-including development of multicellular organisms-and the multiple interactions and feedbacks between DNA, organism, and environment at various levels and timescales in the evolutionary process. First, during the construction of an individual's phenotype, DNA is recruited as a template for building blocks within the cellular context and may in addition be involved in dynamical feedback loops that depend on the environmental and organismal context. Second, in the production of phenotypic variation among individuals, stochastic, environmental, genetic, and parental sources of variation act jointly. While in controlled laboratory settings, various genetic and environmental factors can be tested one at a time or in various combinations, they cannot be separated in natural populations because the environment is not controlled and the genotype can rarely be replicated. Third, along generations, genotype and environment each have specific properties concerning the origin of their variation, the hereditary transmission of this variation, and the evolutionary feedbacks. Natural selection acts as a feedback from phenotype and environment to genotype. This review integrates recent results and concrete examples that illustrate these three points. Although some themes are shared with recent calls and claims to a new conceptual framework in evolutionary biology, the viewpoint presented here only means to add flesh to the standard evolutionary synthesis.
Collapse
Affiliation(s)
- M-A Félix
- Institut de Biologie Ecole Normale Supérieure, CNRS, Paris, France.
| |
Collapse
|
87
|
Saberi A, Jamal A, Beets I, Schoofs L, Newmark PA. GPCRs Direct Germline Development and Somatic Gonad Function in Planarians. PLoS Biol 2016; 14:e1002457. [PMID: 27163480 PMCID: PMC4862687 DOI: 10.1371/journal.pbio.1002457] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/11/2016] [Indexed: 11/21/2022] Open
Abstract
Planarians display remarkable plasticity in maintenance of their germline, with the ability to develop or dismantle reproductive tissues in response to systemic and environmental cues. Here, we investigated the role of G protein-coupled receptors (GPCRs) in this dynamic germline regulation. By genome-enabled receptor mining, we identified 566 putative planarian GPCRs and classified them into conserved and phylum-specific subfamilies. We performed a functional screen to identify NPYR-1 as the cognate receptor for NPY-8, a neuropeptide required for sexual maturation and germ cell differentiation. Similar to NPY-8, knockdown of this receptor results in loss of differentiated germ cells and sexual maturity. NPYR-1 is expressed in neuroendocrine cells of the central nervous system and can be activated specifically by NPY-8 in cell-based assays. Additionally, we screened the complement of GPCRs with expression enriched in sexually reproducing planarians, and identified an orphan chemoreceptor family member, ophis, that controls differentiation of germline stem cells (GSCs). ophis is expressed in somatic cells of male and female gonads, as well as in accessory reproductive tissues. We have previously shown that somatic gonadal cells are required for male GSC specification and maintenance in planarians. However, ophis is not essential for GSC specification or maintenance and, therefore, defines a secondary role for planarian gonadal niche cells in promoting GSC differentiation. Our studies uncover the complement of planarian GPCRs and reveal previously unappreciated roles for these receptors in systemic and local (i.e., niche) regulation of germ cell development. Genome-wide analysis of the planarian Schmidtea mediterranea reveals a complement of over 550 G protein-coupled receptors, including two with critical roles in germline development. G protein-coupled receptors (GPCRs) are the largest and most versatile family of cell-surface receptors. They play critical roles in various cellular and physiological systems and have emerged as a leading group of therapeutic targets. Due to their structural and functional conservation across animals, much has been learned about GPCRs from studies in laboratory models. Here, we performed genome-wide receptor mining to identify and categorize the complement of GPCR-encoding genes in the planarian Schmidtea mediterranea, an emerging model organism for regeneration and germ cell biology. We then conducted two studies implicating planarian GPCRs in the regulation of reproductive function. First, we found the receptor component of a central neuropeptide Y signaling pathway and demonstrated its involvement in the systemic control of reproductive development. Next, we showed that a novel chemoreceptor family member is expressed in somatic cells of the planarian gonads and directs germ cell maturation via the niche. We predict that future studies on the hundreds of other planarian GPCRs identified in this work will not only help us understand the conserved role of these receptors in various physiological pathways but also pave the way for identification of novel therapeutic targets in parasitic relatives of the planarian.
Collapse
Affiliation(s)
- Amir Saberi
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ayana Jamal
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Isabel Beets
- Department of Biology, Functional Genomics and Proteomics Unit, KU Leuven, Leuven, Belgium
| | - Liliane Schoofs
- Department of Biology, Functional Genomics and Proteomics Unit, KU Leuven, Leuven, Belgium
| | - Phillip A. Newmark
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
88
|
Guo H, La Clair JJ, Masler EP, O'Doherty G, Xing Y. De Novo Asymmetric Synthesis and Biological Analysis of the Daumone Pheromones in Caenorhabditis elegans and in the Soybean Cyst Nematode Heterodera glycines. Tetrahedron 2016; 72:2280-2286. [PMID: 29445247 PMCID: PMC5809136 DOI: 10.1016/j.tet.2016.03.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The de novo asymmetric total syntheses of daumone 1, daumone 3 along with 5 new analogs are described. The key steps of our approach are: the diastereoselective palladium catalyzed glycosylation reaction; the Noyori reduction of 2-acetylfuran and an ynone, which introduce the absolute stereochemistry of the sugar and aglycon portion of daumone; and an Achmatowicz rearrangement, an epoxidation and a ring opening installing the remaining asymmetry of daumone. The synthetic daumones 1 and 3 as well as related analogs were evaluated for dauer activity in C. elegans and for effects on hatching of the related nematode H. glycines. This data provides additional structure activity relationships (SAR) that further inform the study of nematode signaling.
Collapse
Affiliation(s)
- Haibing Guo
- School of Nuclear Technology and Chemistry & Life Science, Hubei University of Science and Technology, Xianning 437100, China
| | - James J La Clair
- Xenobe Research Institute, P. O. Box 3052, San Diego, CA 92163-1052, USA
| | - Edward P Masler
- Nematology Laboratory, USDA-ARS-NEA, Beltsville MD 20705, USA
| | - George O'Doherty
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, USA
| | - Yalan Xing
- Department of Chemistry, William Paterson University, 300 Pompton Rd, Wayne, NJ 07470, USA
| |
Collapse
|
89
|
Neal SJ, Park J, DiTirro D, Yoon J, Shibuya M, Choi W, Schroeder FC, Butcher RA, Kim K, Sengupta P. A Forward Genetic Screen for Molecules Involved in Pheromone-Induced Dauer Formation in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2016; 6:1475-87. [PMID: 26976437 PMCID: PMC4856098 DOI: 10.1534/g3.115.026450] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/07/2016] [Indexed: 01/09/2023]
Abstract
Animals must constantly assess their surroundings and integrate sensory cues to make appropriate behavioral and developmental decisions. Pheromones produced by conspecific individuals provide critical information regarding environmental conditions. Ascaroside pheromone concentration and composition are instructive in the decision of Caenorhabditis elegans to either develop into a reproductive adult or enter into the stress-resistant alternate dauer developmental stage. Pheromones are sensed by a small set of sensory neurons, and integrated with additional environmental cues, to regulate neuroendocrine signaling and dauer formation. To identify molecules required for pheromone-induced dauer formation, we performed an unbiased forward genetic screen and identified phd (pheromone response-defective dauer) mutants. Here, we describe new roles in dauer formation for previously identified neuronal molecules such as the WD40 domain protein QUI-1 and MACO-1 Macoilin, report new roles for nociceptive neurons in modulating pheromone-induced dauer formation, and identify tau tubulin kinases as new genes involved in dauer formation. Thus, phd mutants define loci required for the detection, transmission, or integration of pheromone signals in the regulation of dauer formation.
Collapse
Affiliation(s)
- Scott J Neal
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454 National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454
| | - JiSoo Park
- Department of Brain and Cognitive Sciences, DGIST, Daegu 711-873, Republic of Korea
| | - Danielle DiTirro
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454 National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454
| | - Jason Yoon
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454 National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454
| | - Mayumi Shibuya
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454 National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454
| | - Woochan Choi
- Department of Brain and Cognitive Sciences, DGIST, Daegu 711-873, Republic of Korea
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, New York 14853 Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, Florida 32611
| | - Kyuhyung Kim
- Department of Brain and Cognitive Sciences, DGIST, Daegu 711-873, Republic of Korea
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454 National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454
| |
Collapse
|
90
|
cGMP Signalling Mediates Water Sensation (Hydrosensation) and Hydrotaxis in Caenorhabditis elegans. Sci Rep 2016; 6:19779. [PMID: 26891989 PMCID: PMC4759535 DOI: 10.1038/srep19779] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/07/2015] [Indexed: 12/24/2022] Open
Abstract
Animals have developed the ability to sense the water content in their habitats, including hygrosensation (sensing humidity in the air) and hydrosensation (sensing the water content in other microenvironments), and they display preferences for specific water contents that influence their mating, reproduction and geographic distribution. We developed and employed four quantitative behavioural test paradigms to investigate the molecular and cellular mechanisms underlying sensing the water content in an agar substrate (hydrosensation) and hydrotaxis in Caenorhabditis elegans. By combining a reverse genetic screen with genetic manipulation, optogenetic neuronal manipulation and in vivo Ca2+ imaging, we demonstrate that adult worms avoid the wetter areas of agar plates and hypo-osmotic water droplets. We found that the cGMP signalling pathway in ciliated sensory neurons is involved in hydrosensation and hydrotaxis in Caenorhabditis elegans.
Collapse
|
91
|
Sommer RJ, Mayer MG. Toward a Synthesis of Developmental Biology with Evolutionary Theory and Ecology. Annu Rev Cell Dev Biol 2015; 31:453-71. [DOI: 10.1146/annurev-cellbio-102314-112451] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ralf J. Sommer
- Department for Evolutionary Biology, Max-Planck Institute for Developmental Biology, 72076 Tübingen, Germany;
| | - Melanie G. Mayer
- Department for Evolutionary Biology, Max-Planck Institute for Developmental Biology, 72076 Tübingen, Germany;
| |
Collapse
|
92
|
Jensen VL, Li C, Bowie RV, Clarke L, Mohan S, Blacque OE, Leroux MR. Formation of the transition zone by Mks5/Rpgrip1L establishes a ciliary zone of exclusion (CIZE) that compartmentalises ciliary signalling proteins and controls PIP2 ciliary abundance. EMBO J 2015; 34:2537-56. [PMID: 26392567 PMCID: PMC4609185 DOI: 10.15252/embj.201488044] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 08/22/2015] [Accepted: 08/26/2015] [Indexed: 01/09/2023] Open
Abstract
Cilia are thought to harbour a membrane diffusion barrier within their transition zone (TZ) that compartmentalises signalling proteins. How this "ciliary gate" assembles and functions remains largely unknown. Contrary to current models, we present evidence that Caenorhabditis elegans MKS-5 (orthologue of mammalian Mks5/Rpgrip1L/Nphp8 and Rpgrip1) may not be a simple structural scaffold for anchoring > 10 different proteins at the TZ, but instead, functions as an assembly factor. This activity is needed to form TZ ultrastructure, which comprises Y-shaped axoneme-to-membrane connectors. Coiled-coil and C2 domains within MKS-5 enable TZ localisation and functional interactions with two TZ modules, consisting of Meckel syndrome (MKS) and nephronophthisis (NPHP) proteins. Discrete roles for these modules at basal body-associated transition fibres and TZ explain their redundant functions in making essential membrane connections and thus sealing the ciliary compartment. Furthermore, MKS-5 establishes a ciliary zone of exclusion (CIZE) at the TZ that confines signalling proteins, including GPCRs and NPHP-2/inversin, to distal ciliary subdomains. The TZ/CIZE, potentially acting as a lipid gate, limits the abundance of the phosphoinositide PIP2 within cilia and is required for cell signalling. Together, our findings suggest a new model for Mks5/Rpgrip1L in TZ assembly and function that is essential for establishing the ciliary signalling compartment.
Collapse
Affiliation(s)
- Victor L Jensen
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Chunmei Li
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Rachel V Bowie
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lara Clarke
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Swetha Mohan
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
93
|
Rhoads TW, Prasad A, Kwiecien NW, Merrill AE, Zawack K, Westphall MS, Schroeder FC, Kimble J, Coon JJ. NeuCode Labeling in Nematodes: Proteomic and Phosphoproteomic Impact of Ascaroside Treatment in Caenorhabditis elegans. Mol Cell Proteomics 2015; 14:2922-35. [PMID: 26392051 DOI: 10.1074/mcp.m115.049684] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 01/05/2023] Open
Abstract
The nematode Caenorhabditis elegans is an important model organism for biomedical research. We previously described NeuCode stable isotope labeling by amino acids in cell culture (SILAC), a method for accurate proteome quantification with potential for multiplexing beyond the limits of traditional stable isotope labeling by amino acids in cell culture. Here we apply NeuCode SILAC to profile the proteomic and phosphoproteomic response of C. elegans to two potent members of the ascaroside family of nematode pheromones. By consuming labeled E. coli as part of their diet, C. elegans nematodes quickly and easily incorporate the NeuCode heavy lysine isotopologues by the young adult stage. Using this approach, we report, at high confidence, one of the largest proteomic and phosphoproteomic data sets to date in C. elegans: 6596 proteins at a false discovery rate ≤ 1% and 6620 phosphorylation isoforms with localization probability ≥75%. Our data reveal a post-translational signature of pheromone sensing that includes many conserved proteins implicated in longevity and response to stress.
Collapse
Affiliation(s)
| | - Aman Prasad
- ‖Biochemistry, and **Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | | | | | - Kelson Zawack
- ‡‡Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853
| | | | - Frank C Schroeder
- ‡‡Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853
| | - Judith Kimble
- ‖Biochemistry, and **Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Joshua J Coon
- From the Departments of ‡Chemistry, §Biomolecular Chemistry, ¶Genome Center,
| |
Collapse
|
94
|
Neal SJ, Takeishi A, O'Donnell MP, Park J, Hong M, Butcher RA, Kim K, Sengupta P. Feeding state-dependent regulation of developmental plasticity via CaMKI and neuroendocrine signaling. eLife 2015; 4. [PMID: 26335407 PMCID: PMC4558564 DOI: 10.7554/elife.10110] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/31/2015] [Indexed: 01/03/2023] Open
Abstract
Information about nutrient availability is assessed via largely unknown mechanisms to drive developmental decisions, including the choice of Caenorhabditis elegans larvae to enter into the reproductive cycle or the dauer stage. In this study, we show that CMK-1 CaMKI regulates the dauer decision as a function of feeding state. CMK-1 acts cell-autonomously in the ASI, and non cell-autonomously in the AWC, sensory neurons to regulate expression of the growth promoting daf-7 TGF-β and daf-28 insulin-like peptide (ILP) genes, respectively. Feeding state regulates dynamic subcellular localization of CMK-1, and CMK-1-dependent expression of anti-dauer ILP genes, in AWC. A food-regulated balance between anti-dauer ILP signals from AWC and pro-dauer signals regulates neuroendocrine signaling and dauer entry; disruption of this balance in cmk-1 mutants drives inappropriate dauer formation under well-fed conditions. These results identify mechanisms by which nutrient information is integrated in a small neuronal network to modulate neuroendocrine signaling and developmental plasticity. DOI:http://dx.doi.org/10.7554/eLife.10110.001 Living organisms have the remarkable ability to adapt to changes in their external environment. For example, when conditions are favorable, the larvae of the tiny roundworm C. elegans rapidly mature into adults and reproduce. However, when faced with starvation, over-crowding or other adverse conditions, they can stop growing and enter a type of stasis called the dauer stage, which enables them to survive in harsh conditions for extended periods of time. The worms enter the dauer stage if they detect high levels of a pheromone mixture that is produced by other worms—which indicates that the local population is over-crowded. However, temperature, food availability, and other environmental cues also influence this decision. A protein called TGF-β and other proteins called insulin-like peptides are produced by a group of sensory neurons in the worm's head. These proteins usually promote the growth of the worms by increasing the production of particular steroid hormones. However, high levels of the pheromone mixture, an inadequate supply of food and other adverse conditions decrease the expression of the genes that encode these proteins, which allows the worm to enter the dauer state. It is not clear how the worm senses food, nor how this is integrated with the information provided by the pheromones to influence this decision. To address these questions, Neal et al. studied a variety of mutant worms that lacked proteins involved in different aspects of food sensing. The experiments show that worms missing a protein called CaMKI enter the dauer state even under conditions in which food is plentiful and normal worms continue to grow. CaMKI inhibits entry into the dauer stage by increasing the expression of the genes that encode TGF-β and the insulin-like peptides in sensory neurons in response to food. Neal et al.'s findings reveal how CaMKI enables information about food availability to be integrated with other environmental cues to influence whether young worms enter the dauer state. Understanding how food sensing is linked to changes in hormone levels will help us appreciate why and how the availability of food has complex effects on animal biology and behavior. DOI:http://dx.doi.org/10.7554/eLife.10110.002
Collapse
Affiliation(s)
- Scott J Neal
- Department of Biology, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| | - Asuka Takeishi
- Department of Biology, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| | - Michael P O'Donnell
- Department of Biology, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| | - JiSoo Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Myeongjin Hong
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, United States
| | - Kyuhyung Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Piali Sengupta
- Department of Biology, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| |
Collapse
|
95
|
Kim H, Paik YK. Synthesis of Photoaffinity-Labeled Daumone Analogs. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Heekyeong Kim
- Department of Biomaterials Science and Engineering; Yonsei Proteome Research Center
| | - Young-Ki Paik
- Department of Biomaterials Science and Engineering; Yonsei Proteome Research Center
- Department of Integrated Omics for Biomedical Research; Yonsei University; Seoul 120-749 Korea
| |
Collapse
|
96
|
von Reuss SH, Schroeder FC. Combinatorial chemistry in nematodes: modular assembly of primary metabolism-derived building blocks. Nat Prod Rep 2015; 32:994-1006. [PMID: 26059053 PMCID: PMC4884655 DOI: 10.1039/c5np00042d] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The nematode Caenorhabditis elegans was the first animal to have its genome fully sequenced and has become an important model organism for biomedical research. However, like many other animal model systems, its metabolome remained largely uncharacterized, until recent investigations demonstrated the importance of small molecule-based signalling cascades for virtually every aspect of nematode biology. These studies have revealed that nematodes are amazingly skilled chemists: using simple building blocks from conserved primary metabolism and a strategy of modular assembly, C. elegans and other nematode species create complex molecular architectures to regulate their development and behaviour. These nematode-derived modular metabolites (NDMMs) are based on the dideoxysugars ascarylose or paratose, which serve as scaffolds for attachment of moieties from lipid, amino acid, carbohydrate, citrate, and nucleoside metabolism. Mutant screens and comparative metabolomics based on NMR spectroscopy and MS have so-far revealed several 100 different ascarylose ("ascarosides") and a few paratose ("paratosides") derivatives, many of which represent potent signalling molecules that can be active at femtomolar levels, regulating development, behaviour, body shape, and many other life history traits. NDMM biosynthesis appears to be carefully regulated as assembly of different modules proceeds with very high specificity. Preliminary biosynthetic studies have confirmed the primary metabolism origin of some NDMM building blocks, whereas the mechanisms that underlie their highly specific assembly are not understood. Considering their functions and biosynthetic origin, NDMMs represent a new class of natural products that cannot easily be classified as "primary" or "secondary". We believe that the identification of new variants of primary metabolism-derived structures that serve important signalling functions in C. elegans and other nematodes provides a strong incentive for a comprehensive re-analysis of metabolism in higher animals, including humans.
Collapse
Affiliation(s)
- Stephan H. von Reuss
- Max Planck Institute for Chemical Ecology, Department of Bioorganic Chemistry, Jena, Germany
| | - Frank C. Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
97
|
Bi Z, Gong Y, Huang X, Yu H, Bai L, Hu J. Efficacy of Four Nematicides Against the Reproduction and Development of Pinewood Nematode, Bursaphelenchus xylophilus. J Nematol 2015; 47:126-132. [PMID: 26170474 PMCID: PMC4492287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Indexed: 06/04/2023] Open
Abstract
To understand the efficacy of emamectin benzoate, avermectin, milbemectin, and thiacloprid on the reproduction and development of Bursaphelenchus xylophilus, seven parameters, namely population growth, fecundity, egg hatchability, larval lethality, percent larval development, body size, and sexual ratio, were investigated using sublethal (LC20) doses of these compounds in the laboratory. Emamectin benzoate treatment led to a significant suppression in population size, brood size, and percent larval development with 411, 3.50, and 49.63%, respectively, compared to 20850, 24.33, and 61.43% for the negative control. The embryonic and larval lethality increased obviously from 12.47% and 13.70% to 51.37% and 75.30%, respectively. In addition, the body length was also significantly reduced for both males and females in the emamectin benzoate treatment. Avermectin and milbemectin were also effective in suppressing population growth by increasing larval lethality and reducing larval development, although they did not affect either brood size or embryonic lethality. Body length for both male and female worms was increased by avermectin. Thiacloprid caused no adverse reproductive effects, although it suppressed larval development. Sexual ratio was not affected by any of these four nematicides. Our results indicate that emamectin benzoate, milbemectin, and avermectin are effective against the reproduction of B. xylophilus. We think these three nematicides can be useful for the control of pine wilt disease.
Collapse
Affiliation(s)
- Zhenzhen Bi
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China
| | - Yanting Gong
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China
| | - Xiaojuan Huang
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China
| | - Hongshi Yu
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China
| | - Liqun Bai
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China
| | - Jiafu Hu
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang Agricultural & Forestry University, Lin'an, Zhejiang Province, 311300, P.R. China. ; Collaborative Innovation Center of Sustaintable Forestry in Southern China, Nanjing Forestry University
| |
Collapse
|
98
|
Yim JJ, Bose N, Meyer JM, Sommer RJ, Schroeder FC. Nematode signaling molecules derived from multimodular assembly of primary metabolic building blocks. Org Lett 2015; 17:1648-51. [PMID: 25782998 PMCID: PMC4878434 DOI: 10.1021/acs.orglett.5b00329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In the nematode model organisms Caenorhabditis elegans and Pristionchus pacificus, a new class of natural products based on modular assembly of primary-metabolism-derived building blocks control organismal development and behavior. We report identification and biological activities of the first pentamodular metabolite, pasa#9, and the 8-oxoadenine-containing npar#3 from P. pacificus. These structures suggest co-option of nucleoside and tryptophan metabolic pathways for the biosynthesis of endogenous metabolite libraries that transcend the dichotomy between "primary" and "secondary" metabolism.
Collapse
Affiliation(s)
- Joshua J. Yim
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Tubingen 72076, Germany
| | - Neelanjan Bose
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jan M. Meyer
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Tubingen 72076, Germany
| | - Ralf J. Sommer
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Tubingen 72076, Germany
| | - Frank C. Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
99
|
Acyl-CoA oxidase complexes control the chemical message produced by Caenorhabditis elegans. Proc Natl Acad Sci U S A 2015; 112:3955-60. [PMID: 25775534 DOI: 10.1073/pnas.1423951112] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Caenorhabditis elegans uses ascaroside pheromones to induce development of the stress-resistant dauer larval stage and to coordinate various behaviors. Peroxisomal β-oxidation cycles are required for the biosynthesis of the fatty acid-derived side chains of the ascarosides. Here we show that three acyl-CoA oxidases, which catalyze the first step in these β-oxidation cycles, form different protein homo- and heterodimers with distinct substrate preferences. Mutations in the acyl-CoA oxidase genes acox-1, -2, and -3 led to specific defects in ascaroside production. When the acyl-CoA oxidases were expressed alone or in pairs and purified, the resulting acyl-CoA oxidase homo- and heterodimers displayed different side-chain length preferences in an in vitro activity assay. Specifically, an ACOX-1 homodimer controls the production of ascarosides with side chains with nine or fewer carbons, an ACOX-1/ACOX-3 heterodimer controls the production of those with side chains with seven or fewer carbons, and an ACOX-2 homodimer controls the production of those with ω-side chains with less than five carbons. Our results support a biosynthetic model in which β-oxidation enzymes act directly on the CoA-thioesters of ascaroside biosynthetic precursors. Furthermore, we identify environmental conditions, including high temperature and low food availability, that induce the expression of acox-2 and/or acox-3 and lead to corresponding changes in ascaroside production. Thus, our work uncovers an important mechanism by which C. elegans increases the production of the most potent dauer pheromones, those with the shortest side chains, under specific environmental conditions.
Collapse
|
100
|
Meisel JD, Panda O, Mahanti P, Schroeder FC, Kim DH. Chemosensation of bacterial secondary metabolites modulates neuroendocrine signaling and behavior of C. elegans. Cell 2015; 159:267-80. [PMID: 25303524 DOI: 10.1016/j.cell.2014.09.011] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/03/2014] [Accepted: 09/03/2014] [Indexed: 11/30/2022]
Abstract
Discrimination between pathogenic and beneficial microbes is essential for host organism immunity and homeostasis. Here, we show that chemosensory detection of two secondary metabolites produced by Pseudomonas aeruginosa modulates a neuroendocrine signaling pathway that promotes avoidance behavior in the simple animal host Caenorhabditis elegans. Secondary metabolites phenazine-1-carboxamide and pyochelin activate a G-protein-signaling pathway in the ASJ chemosensory neuron pair that induces expression of the neuromodulator DAF-7/TGF-β. DAF-7, in turn, activates a canonical TGF-β signaling pathway in adjacent interneurons to modulate aerotaxis behavior and promote avoidance of pathogenic P. aeruginosa. Our data provide a chemical, genetic, and neuronal basis for how the behavior and physiology of a simple animal host can be modified by the microbial environment and suggest that secondary metabolites produced by microbes may provide environmental cues that contribute to pathogen recognition and host survival.
Collapse
Affiliation(s)
- Joshua D Meisel
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Oishika Panda
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Parag Mahanti
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Dennis H Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|