1
|
Spangler RK, Jonnalagadda K, Ward JD, Partch CL. A wrinkle in timers: evolutionary rewiring of conserved biological timekeepers. Trends Biochem Sci 2025; 50:344-355. [PMID: 39952882 PMCID: PMC12105198 DOI: 10.1016/j.tibs.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Biological timing mechanisms are intrinsic to all organisms, orchestrating the temporal coordination of biological events through complex genetic networks. Circadian rhythms and developmental timers utilize distinct timekeeping mechanisms. This review summarizes the molecular basis for circadian rhythms in mammals and Drosophila, and recent work leveraging these clocks to understand temporal regulation in Caenorhabditis elegans development. We describe the evolutionary connections between distinct timing mechanisms and discuss recent insights into the rewiring of core clock components in development. By integrating findings from circadian and developmental studies with biochemical and structural analyses of conserved components, we aim to illuminate the molecular basis of nematode timing mechanisms and highlight broader insights into biological timing across species.
Collapse
Affiliation(s)
- Rebecca K Spangler
- Department of Chemistry and Biochemistry, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Keya Jonnalagadda
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California - Santa Cruz, Santa Cruz, CA 95064, USA; Center for Circadian Biology, University of California - Santa Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, University of California - Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
2
|
Yamakawa S, Hejnol A. Ecdysteroid-dependent molting in tardigrades. Curr Biol 2024; 34:5804-5812.e4. [PMID: 39566498 DOI: 10.1016/j.cub.2024.10.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/07/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Although molting is a defining feature of the most species-rich animal taxa-the Ecdysozoa, including arthropods, tardigrades, nematodes, and others1,2-its evolutionary background remains enigmatic. In pancrustaceans, such as insects and decapods, molting is regulated by the ecdysteroid (Ecd) hormone and its downstream cascade (Figure 1A, see also the text).3,4,5 However, whether Ecd-dependent molting predates the emergence of the arthropods and represents an ancestral machinery in ecdysozoans remains unclear. For example, involvement of the Ecd hormone in molting regulation has been suggested only in some parasitic nematodes outside of arthropods,6,7 and insect Ecd synthesis and receptor genes are lacking in some ecysozoan lineages (Figure S1A).8,9,10 In this study, we investigated the role of Ecd in the molting process of the tardigrade Hypsibius exemplaris. We show that the endogenous Ecd level periodically increases during the molting cycle of H. exemplaris. The pulse treatment with exogenous Ecd induced molting, whereas an antagonist of the Ecd receptor suppressed the molting. Our spatial and temporal gene expression analysis revealed the putative regulatory organs and Ecd downstream cascades. We demonstrate that tardigrade molting is regulated by the Ecd hormone, supporting the ancestry of Ecd-dependent molting in panarthropods. Furthermore, we were able to identify the putative neural center of molting regulation in tardigrades. This region may be homologous to the neural center in the protocerebrum of pancrustaceans and represent an ancestral state of panarthropods. Together, our results suggest that Ecd-dependent molting evolved in the early-late Ediacaran, 22-76 million years earlier than previously suggested.11.
Collapse
Affiliation(s)
- Shumpei Yamakawa
- Institute of Zoology and Evolutionary Research, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany.
| | - Andreas Hejnol
- Institute of Zoology and Evolutionary Research, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany; Department of Biological Sciences, University of Bergen, 5006 Bergen, Norway.
| |
Collapse
|
3
|
Spangler RK, Braun K, Ashley GE, van der Does M, Wruck D, Coronado AR, Matthew Ragle J, Iesmantavicius V, Morales Moya LJ, Jonnalagadda K, Partch CL, Großhans H, Ward JD. A conserved chronobiological complex times C. elegans development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593322. [PMID: 38766223 PMCID: PMC11100808 DOI: 10.1101/2024.05.09.593322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The mammalian PAS-domain protein PERIOD (PER) and its C. elegans orthologue LIN-42 have been proposed to constitute an evolutionary link between two distinct, circadian and developmental, timing systems. However, while the function of PER in animal circadian rhythms is well understood molecularly and mechanistically, this is not true for LIN-42's function in timing rhythmic development. Here, using targeted deletions, we find that the LIN-42 PAS domains are dispensable for the protein's function in timing molts. Instead, we observe arrhythmic molts upon deletion of a distinct sequence element, conserved with PER. We show that this element, designated CK1δ-binding domain (CK1BD), mediates stable binding to KIN-20, the C. elegans CK1δ/ε orthologue. We demonstrate that CK1δ phosphorylates LIN-42 and define two conserved helical motifs in the CK1BD, CK1BD-A and CK1BD-B, that have distinct roles in controlling CK1δ-binding and kinase activity in vitro. KIN-20 and the LIN-42 CK1BD are required for proper molting timing in vivo, and loss of LIN-42 binding changes KIN-20 subcellular localization. The interactions mirror the central role of a stable circadian PER-CK1 complex in setting a robust ~24-hour period. Hence, our results establish LIN-42/PER - KIN-20/CK1δ/ε as a functionally conserved signaling module of two distinct chronobiological systems.
Collapse
Affiliation(s)
- Rebecca K Spangler
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kathrin Braun
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | - Guinevere E Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Marit van der Does
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Daniel Wruck
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrea Ramos Coronado
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | | | | | - Keya Jonnalagadda
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
- Center for Circadian Biology, University of California-San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California-Santa Cruz, Santa Cruz 95064, USA
| | - Helge Großhans
- Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
4
|
Sundaram MV, Pujol N. The Caenorhabditis elegans cuticle and precuticle: a model for studying dynamic apical extracellular matrices in vivo. Genetics 2024; 227:iyae072. [PMID: 38995735 PMCID: PMC11304992 DOI: 10.1093/genetics/iyae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/25/2024] [Indexed: 07/14/2024] Open
Abstract
Apical extracellular matrices (aECMs) coat the exposed surfaces of animal bodies to shape tissues, influence social interactions, and protect against pathogens and other environmental challenges. In the nematode Caenorhabditis elegans, collagenous cuticle and zona pellucida protein-rich precuticle aECMs alternately coat external epithelia across the molt cycle and play many important roles in the worm's development, behavior, and physiology. Both these types of aECMs contain many matrix proteins related to those in vertebrates, as well as some that are nematode-specific. Extensive differences observed among tissues and life stages demonstrate that aECMs are a major feature of epithelial cell identity. In addition to forming discrete layers, some cuticle components assemble into complex substructures such as ridges, furrows, and nanoscale pillars. The epidermis and cuticle are mechanically linked, allowing the epidermis to sense cuticle damage and induce protective innate immune and stress responses. The C. elegans model, with its optical transparency, facilitates the study of aECM cell biology and structure/function relationships and all the myriad ways by which aECM can influence an organism.
Collapse
Affiliation(s)
- Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nathalie Pujol
- Aix Marseille University, INSERM, CNRS, CIML, Turing Centre for Living Systems, 13009 Marseille, France
| |
Collapse
|
5
|
Patil G, van Zon JS. Timers, variability, and body-wide coordination: C. elegans as a model system for whole-animal developmental timing. Curr Opin Genet Dev 2024; 85:102172. [PMID: 38432125 DOI: 10.1016/j.gde.2024.102172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Successful development requires both precise timing of cellular processes, such as division and differentiation, and tight coordination of timing between tissues and organs. Yet, how time information is encoded with high precision and synchronized between tissues, despite inherent molecular noise, is unsolved. Here, we propose the nematode C. elegans as a unique model system for studying body-wide control of developmental timing. Recent studies combining genetics, quantitative analysis, and simulations have 1) mapped core timers controlling larval development, indicating temporal gradients as an underlying mechanism, and 2) elucidated general principles that make timing insensitive to inherent fluctuations and variation in environmental conditions. As the molecular regulators of C. elegans developmental timing are broadly conserved, these mechanisms likely apply also to higher organisms.
Collapse
|
6
|
Hiroki S, Yoshitane H. Ror homolog nhr-23 is essential for both developmental clock and circadian clock in C. elegans. Commun Biol 2024; 7:243. [PMID: 38418700 PMCID: PMC10902330 DOI: 10.1038/s42003-024-05894-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
Animals have internal clocks that generate biological rhythms. In mammals, clock genes such as Period form the circadian clock to generate approximately 24-h biological rhythms. In C. elegans, the clock gene homologs constitute the "developmental clock", which has an 8-h period during larval development to determine the timing of molting. Thus, the ancestral circadian clock has been believed to evolve into the oscillator with a shorter period in C. elegans. However, circadian rhythms have also been observed in adult C. elegans, albeit relatively weak. This prompts the question: if the clock gene homologs drive the developmental rhythm with 8-h period, which genes generate the circadian rhythms in C. elegans? In this study, we discovered that nhr-23, a homolog of the mammalian circadian clock gene Ror, is essential for circadian transcriptional rhythms in adult C. elegans. Interestingly, nhr-23 was also known to be essential for the molting clock. The bilaterian ancestral circadian clock genes might have evolved to function over multiple periods depending on developmental contexts rather than a single 8-h period in C. elegans.
Collapse
Affiliation(s)
- Shingo Hiroki
- Tokyo Metropolitan Institute of Medical Sciences, Tokyo, Japan.
| | - Hikari Yoshitane
- Tokyo Metropolitan Institute of Medical Sciences, Tokyo, Japan.
- Department of Biological Sciences, School of Science, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
7
|
Hashimshony T, Levin L, Fröbius AC, Dahan N, Chalifa-Caspi V, Hamo R, Gabai-Almog O, Blais I, Assaraf YG, Lubzens E. A transcriptomic examination of encased rotifer embryos reveals the developmental trajectory leading to long-term dormancy; are they "animal seeds"? BMC Genomics 2024; 25:119. [PMID: 38281016 PMCID: PMC10821554 DOI: 10.1186/s12864-024-09961-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Organisms from many distinct evolutionary lineages acquired the capacity to enter a dormant state in response to environmental conditions incompatible with maintaining normal life activities. Most studied organisms exhibit seasonal or annual episodes of dormancy, but numerous less studied organisms enter long-term dormancy, lasting decades or even centuries. Intriguingly, many planktonic animals produce encased embryos known as resting eggs or cysts that, like plant seeds, may remain dormant for decades. Herein, we studied a rotifer Brachionus plicatilis as a model planktonic species that forms encased dormant embryos via sexual reproduction and non-dormant embryos via asexual reproduction and raised the following questions: Which genes are expressed at which time points during embryogenesis? How do temporal transcript abundance profiles differ between the two types of embryos? When does the cell cycle arrest? How do dormant embryos manage energy? RESULTS As the molecular developmental kinetics of encased embryos remain unknown, we employed single embryo RNA sequencing (CEL-seq) of samples collected during dormant and non-dormant embryogenesis. We identified comprehensive and temporal transcript abundance patterns of genes and their associated enriched functional pathways. Striking differences were uncovered between dormant and non-dormant embryos. In early development, the cell cycle-associated pathways were enriched in both embryo types but terminated with fewer nuclei in dormant embryos. As development progressed, the gene transcript abundance profiles became increasingly divergent between dormant and non-dormant embryos. Organogenesis was suspended in dormant embryos, concomitant with low transcript abundance of homeobox genes, and was replaced with an ATP-poor preparatory phase characterized by very high transcript abundance of genes encoding for hallmark dormancy proteins (e.g., LEA proteins, sHSP, and anti-ROS proteins, also found in plant seeds) and proteins involved in dormancy exit. Surprisingly, this period appeared analogous to the late maturation phase of plant seeds. CONCLUSIONS The study highlights novel divergent temporal transcript abundance patterns between dormant and non-dormant embryos. Remarkably, several convergent functional solutions appear during the development of resting eggs and plant seeds, suggesting a similar preparatory phase for long-term dormancy. This study accentuated the broad novel molecular features of long-term dormancy in encased animal embryos that behave like "animal seeds".
Collapse
Affiliation(s)
- Tamar Hashimshony
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Liron Levin
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Andreas C Fröbius
- Molecular Andrology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Gießen, Gießen, Germany.
| | - Nitsan Dahan
- Interdisciplinary Center for Life Sciences and Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Vered Chalifa-Caspi
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Reini Hamo
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Oshri Gabai-Almog
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Idit Blais
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and IVF, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Esther Lubzens
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
- (Retired) Israel Oceanographic and Limnological Research, Haifa, Israel.
| |
Collapse
|
8
|
Lemieux GA, Yoo S, Lin L, Vohra M, Ashrafi K. The steroid hormone ADIOL promotes learning by reducing neural kynurenic acid levels. Genes Dev 2023; 37:998-1016. [PMID: 38092521 PMCID: PMC10760639 DOI: 10.1101/gad.350745.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023]
Abstract
Reductions in brain kynurenic acid levels, a neuroinhibitory metabolite, improve cognitive function in diverse organisms. Thus, modulation of kynurenic acid levels is thought to have therapeutic potential in a range of brain disorders. Here we report that the steroid 5-androstene 3β, 17β-diol (ADIOL) reduces kynurenic acid levels and promotes associative learning in Caenorhabditis elegans We identify the molecular mechanisms through which ADIOL links peripheral metabolic pathways to neural mechanisms of learning capacity. Moreover, we show that in aged animals, which normally experience rapid cognitive decline, ADIOL improves learning capacity. The molecular mechanisms that underlie the biosynthesis of ADIOL as well as those through which it promotes kynurenic acid reduction are conserved in mammals. Thus, rather than a minor intermediate in the production of sex steroids, ADIOL is an endogenous hormone that potently regulates learning capacity by causing reductions in neural kynurenic acid levels.
Collapse
Affiliation(s)
- George A Lemieux
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Shinja Yoo
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lin Lin
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Mihir Vohra
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
9
|
Kinney B, Sahu S, Stec N, Hills-Muckey K, Adams DW, Wang J, Jaremko M, Joshua-Tor L, Keil W, Hammell CM. A circadian-like gene network programs the timing and dosage of heterochronic miRNA transcription during C. elegans development. Dev Cell 2023; 58:2563-2579.e8. [PMID: 37643611 PMCID: PMC10840721 DOI: 10.1016/j.devcel.2023.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/10/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Development relies on the exquisite control of both the timing and the levels of gene expression to achieve robust developmental transitions. How cis- and trans-acting factors control both aspects simultaneously is unclear. We show that transcriptional pulses of the temporal patterning microRNA (miRNA) lin-4 are generated by two nuclear hormone receptors (NHRs) in C. elegans, NHR-85 and NHR-23, whose mammalian orthologs, Rev-Erb and ROR, function in the circadian clock. Although Rev-Erb and ROR antagonize each other to control once-daily transcription in mammals, NHR-85/NHR-23 heterodimers bind cooperatively to lin-4 regulatory elements to induce a single pulse of expression during each larval stage. Each pulse's timing, amplitude, and duration are dictated by the phased expression of these NHRs and the C. elegans Period ortholog, LIN-42, that binds to and represses NHR-85. Therefore, during nematode temporal patterning, an evolutionary rewiring of circadian clock components couples the timing of gene expression to the control of transcriptional dosage.
Collapse
Affiliation(s)
- Brian Kinney
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Shubham Sahu
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168 Laboratoire Physico Chimie Curie, Paris 75005, France
| | - Natalia Stec
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Dexter W Adams
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jing Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Matt Jaremko
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Leemor Joshua-Tor
- Howard Hughes Medical Institute, W. M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wolfgang Keil
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168 Laboratoire Physico Chimie Curie, Paris 75005, France.
| | | |
Collapse
|
10
|
Myles KM, Clancy JC, Johnson LC, Ashley G, Manzano J, Ragle JM, Ward JD. An nhr-85::GFP::AID*::3xFLAG knock-in allele for investigation of molting and oscillatory gene regulation. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000993. [PMID: 37927911 PMCID: PMC10620605 DOI: 10.17912/micropub.biology.000993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023]
Abstract
C. elegans NHR-85 is a poorly characterized nuclear hormone receptor transcription factor with an emerging role in regulating microRNA expression to control developmental timing. We generated the first NHR-85 translational fusion by knocking a GFP::AID*::3xFLAG cassette into the endogenous locus to tag all known isoforms. nhr-85 ::GFP::AID*::3xFLAG animals have wild-type broodsizes and NHR-85 ::GFP peaks in expression at the start of the L4 stage in epithelial cells. NHR-85 is not expressed in the germline, suggesting that while it might cooperate with the NHR-23 transcription factor to control microRNA expression, NHR-23 promotes spermatogenesis independent of NHR-85 . This nhr-85 ::GFP::AID*::3xFLAG strain will be a valuable resource for studying when and where NHR-85 acts to promote developmental timing.
Collapse
Affiliation(s)
- Krista M. Myles
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - John C. Clancy
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - Londen C. Johnson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - Guinevere Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - Jesus Manzano
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - Jordan D. Ward
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| |
Collapse
|
11
|
Myles KM, Ragle JM, Ward JD. An nhr-23::mScarlet::3xMyc knock-in allele for studying spermatogenesis and molting. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000996. [PMID: 37854098 PMCID: PMC10580079 DOI: 10.17912/micropub.biology.000996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
C. elegans NHR-23 is a nuclear hormone receptor transcription factor involved in molting, apical extracellular matrix structure, and spermatogenesis. To determine NHR-23 expression dynamics, we previously tagged the endogenous nhr-23 locus with a GFP::AID*::3xFLAG tag. To allow co-localization of NHR-23 with green fluorescent protein-tagged factors of interest, we generated an equivalent strain carrying an mScarlet::3xMyc tag to produce a C-terminal fusion. Similar to the GFP::AID*::3xFLAG knock-in, NHR-23 ::mScarlet::3xMyc was expressed in seam and hypodermal cells, vulval precursor cells, and the spermatogenic germline. We also observed a diffuse NHR-23::mScarlet expression pattern in spermatids and residual bodies after NHR-23 ceased to localize on chromatin. Further examination of this novel localization may provide insight into NHR-23 regulation of spermatogenesis.
Collapse
Affiliation(s)
- Krista M. Myles
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Jordan D. Ward
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| |
Collapse
|
12
|
Johnson LC, Vo AA, Clancy JC, Myles KM, Pooranachithra M, Aguilera J, Levenson MT, Wohlenberg C, Rechtsteiner A, Ragle JM, Chisholm AD, Ward JD. NHR-23 activity is necessary for C. elegans developmental progression and apical extracellular matrix structure and function. Development 2023; 150:dev201085. [PMID: 37129010 PMCID: PMC10233720 DOI: 10.1242/dev.201085] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 04/11/2023] [Indexed: 05/03/2023]
Abstract
Nematode molting is a remarkable process where animals must repeatedly build a new apical extracellular matrix (aECM) beneath a previously built aECM that is subsequently shed. The nuclear hormone receptor NHR-23 (also known as NR1F1) is an important regulator of C. elegans molting. NHR-23 expression oscillates in the epidermal epithelium, and soma-specific NHR-23 depletion causes severe developmental delay and death. Tissue-specific RNAi suggests that nhr-23 acts primarily in seam and hypodermal cells. NHR-23 coordinates the expression of factors involved in molting, lipid transport/metabolism and remodeling of the aECM. NHR-23 depletion causes dampened expression of a nas-37 promoter reporter and a loss of reporter oscillation. The cuticle collagen ROL-6 and zona pellucida protein NOAH-1 display aberrant annular localization and severe disorganization over the seam cells after NHR-23 depletion, while the expression of the adult-specific cuticle collagen BLI-1 is diminished and frequently found in patches. Consistent with these localization defects, the cuticle barrier is severely compromised when NHR-23 is depleted. Together, this work provides insight into how NHR-23 acts in the seam and hypodermal cells to coordinate aECM regeneration during development.
Collapse
Affiliation(s)
- Londen C. Johnson
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - An A. Vo
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - John C. Clancy
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Krista M. Myles
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Murugesan Pooranachithra
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph Aguilera
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Max T. Levenson
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Chloe Wohlenberg
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andreas Rechtsteiner
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrew D. Chisholm
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jordan D. Ward
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
13
|
Meeuse MWM, Hauser YP, Nahar S, Smith AAT, Braun K, Azzi C, Rempfler M, Großhans H. C. elegans molting requires rhythmic accumulation of the Grainyhead/LSF transcription factor GRH-1. EMBO J 2023; 42:e111895. [PMID: 36688410 PMCID: PMC9929640 DOI: 10.15252/embj.2022111895] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 01/24/2023] Open
Abstract
C. elegans develops through four larval stages that are rhythmically terminated by molts, that is, the synthesis and shedding of a cuticular exoskeleton. Each larval cycle involves rhythmic accumulation of thousands of transcripts, which we show here relies on rhythmic transcription. To uncover the responsible gene regulatory networks (GRNs), we screened for transcription factors that promote progression through the larval stages and identified GRH-1, BLMP-1, NHR-23, NHR-25, MYRF-1, and BED-3. We further characterize GRH-1, a Grainyhead/LSF transcription factor, whose orthologues in other animals are key epithelial cell-fate regulators. We find that GRH-1 depletion extends molt durations, impairs cuticle integrity and shedding, and causes larval death. GRH-1 is required for, and accumulates prior to, each molt, and preferentially binds to the promoters of genes expressed during this time window. Binding to the promoters of additional genes identified in our screen furthermore suggests that we have identified components of a core molting-clock GRN. Since the mammalian orthologues of GRH-1, BLMP-1 and NHR-23, have been implicated in rhythmic homeostatic skin regeneration in mouse, the mechanisms underlying rhythmic C. elegans molting may apply beyond nematodes.
Collapse
Affiliation(s)
- Milou W M Meeuse
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
- University of BaselBaselSwitzerland
| | - Yannick P Hauser
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
- University of BaselBaselSwitzerland
| | - Smita Nahar
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
| | | | - Kathrin Braun
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
| | - Chiara Azzi
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
- University of BaselBaselSwitzerland
| | - Markus Rempfler
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
| | - Helge Großhans
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
- University of BaselBaselSwitzerland
| |
Collapse
|
14
|
Liu M, Gao X, Shan S, Li Y, Wang J, Lu W. Eleutheroside E reduces intestinal fat accumulation in Caenorhabditis elegans through neuroendocrine signals. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:5219-5228. [PMID: 35297055 DOI: 10.1002/jsfa.11875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/13/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Acanthopanax senticosus, a small woody shrub of the family Araliaceae, can be used as a functional food with multiple biological activities. Eleutheroside E (EE), an important active component of A. senticosus, has significant effects on neurological diseases. However, whether EE can regulate lipid metabolism has not been reported. The brain can mediate communication between neurons and intestinal cells through long-distance neuroendocrine signals. We speculated that EE might regulate the intestinal lipid metabolism of Caenorhabditis elegans through neuroendocrine signals. RESULTS First, we found that EE reduced the intestinal fat content of C. elegans, without affecting development, reproduction, food intake or movement. In addition, EE significantly regulated genes and metabolites related to lipid metabolism. EE extensively affected fatty acid synthesis, β-oxidation and lipolysis processes, and regulated the content of various fatty acid and lipid metabolism intermediates. We finally proved that EE reduced intestinal fat storage through serotonin and neuropeptide flp-7-npr-22 pathways in the nervous system. CONCLUSION EE is expected to be a functional food that regulates lipid metabolism. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Mengyao Liu
- Institute of Extreme Environment Nutrition and Protection, Harbin Institute of Technology, Harbin, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, China
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Xin Gao
- Institute of Extreme Environment Nutrition and Protection, Harbin Institute of Technology, Harbin, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, China
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Shan Shan
- Institute of Extreme Environment Nutrition and Protection, Harbin Institute of Technology, Harbin, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, China
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Yongzhi Li
- Key Laboratory of Astronaut Health Center, China Astronaut Research and Training Center, Beijing, China
| | - Jiaping Wang
- Key Laboratory of Astronaut Health Center, China Astronaut Research and Training Center, Beijing, China
| | - Weihong Lu
- Institute of Extreme Environment Nutrition and Protection, Harbin Institute of Technology, Harbin, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, China
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
15
|
Joseph BB, Edeen PT, Meadows S, Binti S, Fay DS. An unexpected role for the conserved ADAM-family metalloprotease ADM-2 in Caenorhabditis elegans molting. PLoS Genet 2022; 18:e1010249. [PMID: 35639786 PMCID: PMC9187072 DOI: 10.1371/journal.pgen.1010249] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/10/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Molting is a widespread developmental process in which the external extracellular matrix (ECM), the cuticle, is remodeled to allow for organismal growth and environmental adaptation. Studies in the nematode Caenorhabditis elegans have identified a diverse set of molting-associated factors including signaling molecules, intracellular trafficking regulators, ECM components, and ECM-modifying enzymes such as matrix metalloproteases. C. elegans NEKL-2 and NEKL-3, two conserved members of the NEK family of protein kinases, are essential for molting and promote the endocytosis of environmental steroid-hormone precursors by the epidermis. Steroids in turn drive the cyclic induction of many genes required for molting. Here we report a role for the sole C. elegans ADAM–meltrin metalloprotease family member, ADM-2, as a mediator of molting. Loss of adm-2, including mutations that disrupt the metalloprotease domain, led to the strong suppression of molting defects in partial loss-of-function nekl mutants. ADM-2 is expressed in the epidermis, and its trafficking through the endo-lysosomal network was disrupted after NEKL depletion. We identified the epidermally expressed low-density lipoprotein receptor–related protein, LRP-1, as a candidate target of ADM-2 regulation. Whereas loss of ADM-2 activity led to the upregulation of apical epidermal LRP-1, ADM-2 overexpression caused a reduction in LRP-1 levels. Consistent with this, several mammalian ADAMs, including the meltrin ADAM12, have been shown to regulate mammalian LRP1 via proteolysis. In contrast to mammalian homologs, however, the regulation of LRP-1 by ADM-2 does not appear to involve the metalloprotease function of ADM-2, nor is proteolytic processing of LRP-1 strongly affected in adm-2 mutants. Our findings suggest a noncanonical role for an ADAM family member in the regulation of a lipoprotein-like receptor and lead us to propose that endocytic trafficking may be important for both the internalization of factors that promote molting as well as the removal of proteins that can inhibit the process. The molecular and cellular features of molting in nematodes share many similarities with cellular and developmental processes that occur in mammals. This includes the degradation and reorganization of extracellular matrix materials that surround cells, as well as the intracellular machineries that allow cells to sample and modify their environments. In the current study, we found an unexpected function for a conserved protein that cleaves other proteins on the external surface of cells. Rather than promoting molting through extracellular matrix reorganization, however, the ADM-2 protease appears to function as a negative regulator of molting. This observation can be explained in part by data showing that ADM-2 negatively regulates a cell surface receptor required for molting. Surprisingly, it appears to do so through a mechanism that does not involve proteolysis. Our data provide insights into the mechanisms controlling molting and link several conserved proteins to show how they function together during development.
Collapse
Affiliation(s)
- Braveen B. Joseph
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Phillip T. Edeen
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Sarina Meadows
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Shaonil Binti
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
- * E-mail:
| |
Collapse
|
16
|
Kalia V, Niedzwiecki MM, Bradner JM, Lau FK, Anderson FL, Bucher ML, Manz KE, Schlotter AP, Fuentes ZC, Pennell KD, Picard M, Walker DI, Hu WT, Jones DP, Miller GW. Cross-species metabolomic analysis of tau- and DDT-related toxicity. PNAS NEXUS 2022; 1:pgac050. [PMID: 35707205 PMCID: PMC9186048 DOI: 10.1093/pnasnexus/pgac050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/28/2022] [Indexed: 01/29/2023]
Abstract
Exposure to the pesticide dichlorodiphenyltrichloroethane (DDT) has been associated with increased risk of Alzheimer's disease (AD), a disease also associated with hyperphosphorylated tau (p-tau) protein aggregation. We investigated whether exposure to DDT can exacerbate tau protein toxicity in Caenorhabditiselegans using a transgenic strain that expresses human tau protein prone to aggregation by measuring changes in size, swim behavior, respiration, lifespan, learning, and metabolism. In addition, we examined the association between cerebrospinal fluid (CSF) p-tau protein-as a marker of postmortem tau burden-and global metabolism in both a human population study and in C. elegans, using the same p-tau transgenic strain. From the human population study, plasma and CSF-derived metabolic features associated with p-tau levels were related to drug, amino acid, fatty acid, and mitochondrial metabolism pathways. A total of five metabolites overlapped between plasma and C. elegans, and four between CSF and C. elegans. DDT exacerbated the inhibitory effect of p-tau protein on growth and basal respiration. In the presence of p-tau protein, DDT induced more curling and was associated with reduced levels of amino acids but increased levels of uric acid and adenosylselenohomocysteine. Our findings in C. elegans indicate that DDT exposure and p-tau aggregation both inhibit mitochondrial function and DDT exposure can exacerbate the mitochondrial inhibitory effects of p-tau aggregation. Further, biological pathways associated with exposure to DDT and p-tau protein appear to be conserved between species.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Megan M Niedzwiecki
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Fion K Lau
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Faith L Anderson
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Katherine E Manz
- School of Engineering, Brown University, Providence, RI, 02912 USA
| | - Alexa Puri Schlotter
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| | - Zoe Coates Fuentes
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Kurt D Pennell
- School of Engineering, Brown University, Providence, RI, 02912 USA
| | - Martin Picard
- Department of Neurology, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032 USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - William T Hu
- Department of Neurology, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901 USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, 30322 USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032 USA
| |
Collapse
|
17
|
Doering KRS, Cheng X, Milburn L, Ratnappan R, Ghazi A, Miller DL, Taubert S. Nuclear hormone receptor NHR-49 acts in parallel with HIF-1 to promote hypoxia adaptation in Caenorhabditis elegans. eLife 2022; 11:e67911. [PMID: 35285794 PMCID: PMC8959602 DOI: 10.7554/elife.67911] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/12/2022] [Indexed: 01/06/2023] Open
Abstract
The response to insufficient oxygen (hypoxia) is orchestrated by the conserved hypoxia-inducible factor (HIF). However, HIF-independent hypoxia response pathways exist that act in parallel with HIF to mediate the physiological hypoxia response. Here, we describe a hypoxia response pathway controlled by Caenorhabditis elegans nuclear hormone receptor NHR-49, an orthologue of mammalian peroxisome proliferator-activated receptor alpha (PPARα). We show that nhr-49 is required for animal survival in hypoxia and is synthetic lethal with hif-1 in this context, demonstrating that these factors act in parallel. RNA-seq analysis shows that in hypoxia nhr-49 regulates a set of genes that are hif-1-independent, including autophagy genes that promote hypoxia survival. We further show that nuclear hormone receptor nhr-67 is a negative regulator and homeodomain-interacting protein kinase hpk-1 is a positive regulator of the NHR-49 pathway. Together, our experiments define a new, essential hypoxia response pathway that acts in parallel with the well-known HIF-mediated hypoxia response.
Collapse
Affiliation(s)
- Kelsie RS Doering
- Graduate Program in Medical Genetics, University of British ColumbiaVancouverCanada
- British Columbia Children's Hospital Research InstituteVancouverCanada
- Centre for Molecular Medicine and Therapeutics, The University of British ColumbiaVancouverCanada
| | - Xuanjin Cheng
- British Columbia Children's Hospital Research InstituteVancouverCanada
- Centre for Molecular Medicine and Therapeutics, The University of British ColumbiaVancouverCanada
- Department of Medical Genetics, University of British ColumbiaVancouverCanada
| | - Luke Milburn
- Department of Biochemistry, University of Washington School of MedicineSeattleUnited States
| | - Ramesh Ratnappan
- Department of Pediatrics, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Arjumand Ghazi
- Department of Pediatrics, University of Pittsburgh School of MedicinePittsburghUnited States
- Departments of Developmental Biology and Cell Biology and Physiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Dana L Miller
- Department of Biochemistry, University of Washington School of MedicineSeattleUnited States
| | - Stefan Taubert
- Graduate Program in Medical Genetics, University of British ColumbiaVancouverCanada
- British Columbia Children's Hospital Research InstituteVancouverCanada
- Centre for Molecular Medicine and Therapeutics, The University of British ColumbiaVancouverCanada
- Department of Medical Genetics, University of British ColumbiaVancouverCanada
| |
Collapse
|
18
|
Taubenheim J, Kortmann C, Fraune S. Function and Evolution of Nuclear Receptors in Environmental-Dependent Postembryonic Development. Front Cell Dev Biol 2021; 9:653792. [PMID: 34178983 PMCID: PMC8222990 DOI: 10.3389/fcell.2021.653792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear receptors (NRs) fulfill key roles in the coordination of postembryonal developmental transitions in animal species. They control the metamorphosis and sexual maturation in virtually all animals and by that the two main environmental-dependent developmental decision points. Sexual maturation and metamorphosis are controlled by steroid receptors and thyroid receptors, respectively in vertebrates, while both processes are orchestrated by the ecdysone receptor (EcR) in insects. The regulation of these processes depends on environmental factors like nutrition, temperature, or photoperiods and by that NRs form evolutionary conserved mediators of phenotypic plasticity. While the mechanism of action for metamorphosis and sexual maturation are well studied in model organisms, the evolution of these systems is not entirely understood and requires further investigation. We here review the current knowledge of NR involvement in metamorphosis and sexual maturation across the animal tree of life with special attention to environmental integration and evolution of the signaling mechanism. Furthermore, we compare commonalities and differences of the different signaling systems. Finally, we identify key gaps in our knowledge of NR evolution, which, if sufficiently investigated, would lead to an importantly improved understanding of the evolution of complex signaling systems, the evolution of life history decision points, and, ultimately, speciation events in the metazoan kingdom.
Collapse
Affiliation(s)
| | | | - Sebastian Fraune
- Zoology and Organismic Interactions, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Transcriptional profiles in Strongyloides stercoralis males reveal deviations from the Caenorhabditis sex determination model. Sci Rep 2021; 11:8254. [PMID: 33859232 PMCID: PMC8050236 DOI: 10.1038/s41598-021-87478-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/30/2021] [Indexed: 02/02/2023] Open
Abstract
The human and canine parasitic nematode Strongyloides stercoralis utilizes an XX/XO sex determination system, with parasitic females reproducing by mitotic parthenogenesis and free-living males and females reproducing sexually. However, the genes controlling S. stercoralis sex determination and male development are unknown. We observed precocious development of rhabditiform males in permissive hosts treated with corticosteroids, suggesting that steroid hormones can regulate male development. To examine differences in transcript abundance between free-living adult males and other developmental stages, we utilized RNA-Seq. We found two clusters of S. stercoralis-specific genes encoding predicted transmembrane proteins that are only expressed in free-living males. We additionally identified homologs of several genes important for sex determination in Caenorhabditis species, including mab-3, tra-1, fem-2, and sex-1, which may have similar functions. However, we identified three paralogs of gld-1; Ss-qki-1 transcripts were highly abundant in adult males, while Ss-qki-2 and Ss-qki-3 transcripts were highly abundant in adult females. We also identified paralogs of pumilio domain-containing proteins with sex-specific transcripts. Intriguingly, her-1 appears to have been lost in several parasite lineages, and we were unable to identify homologs of tra-2 outside of Caenorhabditis species. Together, our data suggest that different mechanisms control male development in S. stercoralis and Caenorhabditis species.
Collapse
|
20
|
Baugh LR, Hu PJ. Starvation Responses Throughout the Caenorhabditiselegans Life Cycle. Genetics 2020; 216:837-878. [PMID: 33268389 PMCID: PMC7768255 DOI: 10.1534/genetics.120.303565] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Caenorhabditis elegans survives on ephemeral food sources in the wild, and the species has a variety of adaptive responses to starvation. These features of its life history make the worm a powerful model for studying developmental, behavioral, and metabolic starvation responses. Starvation resistance is fundamental to life in the wild, and it is relevant to aging and common diseases such as cancer and diabetes. Worms respond to acute starvation at different times in the life cycle by arresting development and altering gene expression and metabolism. They also anticipate starvation during early larval development, engaging an alternative developmental program resulting in dauer diapause. By arresting development, these responses postpone growth and reproduction until feeding resumes. A common set of signaling pathways mediates systemic regulation of development in each context but with important distinctions. Several aspects of behavior, including feeding, foraging, taxis, egg laying, sleep, and associative learning, are also affected by starvation. A variety of conserved signaling, gene regulatory, and metabolic mechanisms support adaptation to starvation. Early life starvation can have persistent effects on adults and their descendants. With its short generation time, C. elegans is an ideal model for studying maternal provisioning, transgenerational epigenetic inheritance, and developmental origins of adult health and disease in humans. This review provides a comprehensive overview of starvation responses throughout the C. elegans life cycle.
Collapse
Affiliation(s)
- L Ryan Baugh
- Department of Biology, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708 and
| | - Patrick J Hu
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
21
|
Ragle JM, Aita AL, Morrison KN, Martinez-Mendez R, Saeger HN, Ashley GA, Johnson LC, Schubert KA, Shakes DC, Ward JD. The conserved molting/circadian rhythm regulator NHR-23/NR1F1 serves as an essential co-regulator of C. elegans spermatogenesis. Development 2020; 147:dev193862. [PMID: 33060131 PMCID: PMC7710015 DOI: 10.1242/dev.193862] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
In sexually reproducing metazoans, spermatogenesis is the process by which uncommitted germ cells give rise to haploid sperm. Work in model systems has revealed mechanisms controlling commitment to the sperm fate, but how this fate is subsequently executed remains less clear. While studying the well-established role of the conserved nuclear hormone receptor transcription factor, NHR-23/NR1F1, in regulating C. elegans molting, we discovered that NHR-23/NR1F1 is also constitutively expressed in developing primary spermatocytes and is a critical regulator of spermatogenesis. In this novel role, NHR-23/NR1F1 functions downstream of the canonical sex-determination pathway. Degron-mediated depletion of NHR-23/NR1F1 within hermaphrodite or male germlines causes sterility due to an absence of functional sperm, as depleted animals produce arrested primary spermatocytes rather than haploid sperm. These spermatocytes arrest in prometaphase I and fail to either progress to anaphase or attempt spermatid-residual body partitioning. They make sperm-specific membranous organelles but fail to assemble their major sperm protein into fibrous bodies. NHR-23/NR1F1 appears to function independently of the known SPE-44 gene regulatory network, revealing the existence of an NHR-23/NR1F1-mediated module that regulates the spermatogenesis program.
Collapse
Affiliation(s)
- James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Abigail L Aita
- Department of Biology, William & Mary, Williamsburg, VA 23187, USA
| | | | - Raquel Martinez-Mendez
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hannah N Saeger
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Guinevere A Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Londen C Johnson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Katherine A Schubert
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Diane C Shakes
- Department of Biology, William & Mary, Williamsburg, VA 23187, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
22
|
Li Y, Feng Y, Wang X, Cui J, Deng X, Zhang X. Adaptation of pine wood nematode Bursaphelenchus xylophilus to β-pinene stress. BMC Genomics 2020; 21:478. [PMID: 32660425 PMCID: PMC7358211 DOI: 10.1186/s12864-020-06876-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/01/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The pine wood nematode (PWN; Bursaphelenchus xylophilus) is the most damaging biological pest in pine forest ecosystems in China. However, the pathogenic mechanism remains unclear. Tracheid cavitation induced by excess metabolism of volatile terpenes is a typical characteristic of pine trees infected by B. xylophilus. β-pinene, one of the main volatile terpenes, influences PWN colonization and reproduction, stimulating pathogenicity during the early stages of infection. To elucidate the response mechanism of PWN to β-pinene, pathogenesis, mortality, and reproduction rate were investigated under different concentrations of β-pinene using a transcriptomics approach. RESULTS A low concentration of β-pinene (BL, C < 25.74 mg/ml) inhibited PWN reproduction, whereas a high concentration (BH, C > 128.7 mg/ml) promoted reproduction. Comparison of PWN expression profiles under low (BL, 21.66 mg/ml) and high (BH, 214.5 mg/ml) β-pinene concentrations at 48 h identified 659 and 418 differentially expressed genes (DEGs), respectively, compared with controls. Some key DEGs are potential regulators of β-pinene via detoxification metabolism (cytochrome P450, UDP-glucuronosyltransferases and short-chain dehydrogenases), ion channel/transporter activity (unc and ATP-binding cassette families), and nuclear receptor -related genes. Gene Ontology enrichment analysis of DEGs revealed metabolic processes as the most significant biological processes, and catalytic activity as the most significant molecular function for both BL and BH samples. Kyoto Encyclopedia of Genes and Genomes (KEGG) Orthology (KO) analysis showed that xenobiotics biodegradation and metabolism, carbohydrate metabolism, lipid metabolism, amino acid metabolism, metabolism of cofactors and vitamins, and transport and catabolism were the dominant terms in metabolism categories. CONCLUSION In addition to detoxification via reduction/oxidation (redox) activity, PWN responds to β-pinene through amino acid metabolism, carbohydrate metabolism, and other pathways including growth regulation and epidermal protein changes to overcome β-pinene stress. This study lays a foundation for further exploring the pathogenic mechanism of PWN.
Collapse
Affiliation(s)
- Yongxia Li
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| | - Yuqian Feng
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| | - Xuan Wang
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| | - Jing Cui
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| | - Xun Deng
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| | - Xingyao Zhang
- Lab. of Forest Pathogen Integrated Biology, Research institute of Forestry New Technology, Chinese Academy of Forestry, Beijing, 100091 China
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037 China
| |
Collapse
|
23
|
Ewe CK, Torres Cleuren YN, Rothman JH. Evolution and Developmental System Drift in the Endoderm Gene Regulatory Network of Caenorhabditis and Other Nematodes. Front Cell Dev Biol 2020; 8:170. [PMID: 32258041 PMCID: PMC7093329 DOI: 10.3389/fcell.2020.00170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/02/2020] [Indexed: 01/17/2023] Open
Abstract
Developmental gene regulatory networks (GRNs) underpin metazoan embryogenesis and have undergone substantial modification to generate the tremendous variety of animal forms present on Earth today. The nematode Caenorhabditis elegans has been a central model for advancing many important discoveries in fundamental mechanistic biology and, more recently, has provided a strong base from which to explore the evolutionary diversification of GRN architecture and developmental processes in other species. In this short review, we will focus on evolutionary diversification of the GRN for the most ancient of the embryonic germ layers, the endoderm. Early embryogenesis diverges considerably across the phylum Nematoda. Notably, while some species deploy regulative development, more derived species, such as C. elegans, exhibit largely mosaic modes of embryogenesis. Despite the relatively similar morphology of the nematode gut across species, widespread variation has been observed in the signaling inputs that initiate the endoderm GRN, an exemplar of developmental system drift (DSD). We will explore how genetic variation in the endoderm GRN helps to drive DSD at both inter- and intraspecies levels, thereby resulting in a robust developmental system. Comparative studies using divergent nematodes promise to unveil the genetic mechanisms controlling developmental plasticity and provide a paradigm for the principles governing evolutionary modification of an embryonic GRN.
Collapse
Affiliation(s)
- Chee Kiang Ewe
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| | | | - Joel H. Rothman
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
24
|
Rashid S, Pho KB, Mesbahi H, MacNeil LT. Nutrient Sensing and Response Drive Developmental Progression in Caenorhabditis elegans. Bioessays 2020; 42:e1900194. [PMID: 32003906 DOI: 10.1002/bies.201900194] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/22/2019] [Indexed: 12/18/2022]
Abstract
In response to nutrient limitation, many animals, including Caenorhabditis elegans, slow or arrest their development. This process requires mechanisms that sense essential nutrients and induce appropriate responses. When faced with nutrient limitation, C. elegans can induce both short and long-term survival strategies, including larval arrest, decreased developmental rate, and dauer formation. To select the most advantageous strategy, information from many different sensors must be integrated into signaling pathways, including target of rapamycin (TOR) and insulin, that regulate developmental progression. Here, how nutrient information is sensed and integrated into developmental decisions that determine developmental rate and progression in C. elegans is reviewed.
Collapse
Affiliation(s)
- Sabih Rashid
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Kim B Pho
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Hiva Mesbahi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| |
Collapse
|
25
|
Bui LT, Ragsdale EJ. Multiple plasticity regulators reveal targets specifying an induced predatory form in nematodes. Mol Biol Evol 2019; 36:2387-2399. [PMID: 31364718 DOI: 10.1093/molbev/msz171] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/19/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022] Open
Abstract
The ability to translate a single genome into multiple phenotypes, or developmental plasticity, defines how phenotype derives from more than just genes. However, to study the evolutionary targets of plasticity and their evolutionary fates, we need to understand how genetic regulators of plasticity control downstream gene expression. Here, we have identified a transcriptional response specific to polyphenism (i.e., discrete plasticity) in the nematode Pristionchus pacificus. This species produces alternative resource-use morphs - microbivorous and predatory forms, differing in the form of their teeth, a morphological novelty - as influenced by resource availability. Transcriptional profiles common to multiple polyphenism-controlling genes in P. pacificus reveal a suite of environmentally sensitive loci, or ultimate target genes, that make up an induced developmental response. Additionally, in vitro assays show that one polyphenism regulator, the nuclear receptor (NR) NHR-40, physically binds to promoters with putative HNF4⍺ (the NR class including NHR-40) binding sites, suggesting this receptor may directly regulate genes that describe alternative morphs. Among differentially expressed genes were morph-limited genes, highlighting factors with putative "on-off" function in plasticity regulation. Further, predatory morph-biased genes included candidates - namely, all four P. pacificus homologs of Hsp70, which have HNF4⍺ motifs - whose natural variation in expression matches phenotypic differences among P. pacificus wild isolates. In summary, our study links polyphenism regulatory loci to the transcription producing alternative forms of a morphological novelty. Consequently, our findings establish a platform for determining how specific regulators of morph-biased genes may influence selection on plastic phenotypes.
Collapse
Affiliation(s)
- Linh T Bui
- Department of Biology, Indiana University, Bloomington, IN
| | | |
Collapse
|
26
|
Pinet K, McLaughlin KA. Mechanisms of physiological tissue remodeling in animals: Manipulating tissue, organ, and organism morphology. Dev Biol 2019; 451:134-145. [DOI: 10.1016/j.ydbio.2019.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/21/2022]
|
27
|
Schumann I, Kenny N, Hui J, Hering L, Mayer G. Halloween genes in panarthropods and the evolution of the early moulting pathway in Ecdysozoa. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180888. [PMID: 30839709 PMCID: PMC6170570 DOI: 10.1098/rsos.180888] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/17/2018] [Indexed: 05/15/2023]
Abstract
Moulting is a characteristic feature of Ecdysozoa-the clade of moulting animals that includes the hyperdiverse arthropods and less speciose groups, such as onychophorans, tardigrades and nematodes. Moulting has been best analysed in arthropods, specifically in insects and crustaceans, in which a complex neuroendocrine system acts at the genomic level and initiates the transcription of genes responsible for moulting. The key moulting hormones, ecdysone and 20-hydroxyecdysone, are subsequently synthesized from cholesterol ingested with food. Their biosynthesis is regulated by the Rieske-domain protein Neverland and cytochrome P450 enzymes encoded by the so-called 'Halloween' genes. Ecdysone is then released into the haemolymph and modified into 20-hydroxyecdysone, which binds to the nuclear receptor EcR/USP and initiates transcription of the Early genes. As little is known about the moulting pathway of other ecdysozoans, we examined the occurrence of genes involved in ecdysteroid biosynthesis and the early moulting cascade across ecdysozoan subgroups. Genomic and transcriptomic searches revealed no Halloween genes in cycloneuralians, whereas only shadow (CYP315A1) is present in onychophorans and tardigrades, suggesting that the Halloween genes evolved stepwise in panarthropods. These findings imply that the genes which were responsible for the ecdysteroid biosynthesis in the last common ancestor of Ecdysozoa are currently unknown.
Collapse
Affiliation(s)
- Isabell Schumann
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
- Molecular Evolution and Animal Systematics, Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Nathan Kenny
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Center of Soybean Research, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, People's Republic of China
| | - Jerome Hui
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Center of Soybean Research, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, People's Republic of China
| | - Lars Hering
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| | - Georg Mayer
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| |
Collapse
|
28
|
Xu X, Zhang YN, Peng S, Wu J, Deng D, Zhou Z. Effects of Microcystis aeruginosa on the expression of nuclear receptor genes in Daphnia similoides sinensis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 150:344-352. [PMID: 29306189 DOI: 10.1016/j.ecoenv.2017.12.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 06/07/2023]
Abstract
Nuclear receptor (NR) genes form a conserved superfamily, which is involved in organism metabolism, reproduction, development, homeostasis, and resource allocation. Microcystis aeruginosa can inhibit the growth and reproduction of Daphnia. However, whether M. aeruginosa can affect the expression of Daphnia NR genes is unknown. In total, 18 NRs were identified in this study based on previous Daphnia similoides sinensis transcriptome data. In treatments containing M. aeruginosa, the gene expression of the NR1 subfamily (E75a, E75b, HR3, HR96, NHR-1, HR97a, HR97g, and NHR97) and the NR2 subfamily (RXR, TLL, PNR, and SVP) were down-regulated 59% and 79%, respectively. In treatments containing M. aeruginosa, although the expression of 78% of the genes showed a similar trend in clones 1 and 2, the expression of 42% of the genes in clone 3 showed the opposite trend compared to clones 1 and 2, suggesting that the adaptability and molecular mechanism differ in individuals with different Microcystis tolerance genotypes.
Collapse
Affiliation(s)
- Xiaoxue Xu
- School of Resources and Environmental Engineering, Anhui University, 230601, Hefei, Anhui, China; School of Life Science, Huaibei Normal University, 235000 Huaibei, Anhui, China
| | - Ya-Nan Zhang
- School of Life Science, Huaibei Normal University, 235000 Huaibei, Anhui, China
| | - Shuixiu Peng
- School of Life Science, Huaibei Normal University, 235000 Huaibei, Anhui, China
| | - Jianxun Wu
- School of Resources and Environmental Engineering, Anhui University, 230601, Hefei, Anhui, China
| | - Daogui Deng
- School of Life Science, Huaibei Normal University, 235000 Huaibei, Anhui, China.
| | - Zhongze Zhou
- School of Resources and Environmental Engineering, Anhui University, 230601, Hefei, Anhui, China.
| |
Collapse
|
29
|
Specific regulation of thermosensitive lipid droplet fusion by a nuclear hormone receptor pathway. Proc Natl Acad Sci U S A 2017; 114:8841-8846. [PMID: 28760992 DOI: 10.1073/pnas.1704277114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Nuclear receptors play important roles in regulating fat metabolism and energy production in humans. The regulatory functions and endogenous ligands of many nuclear receptors are still unidentified, however. Here, we report that CYP-37A1 (ortholog of human cytochrome P450 CYP4V2), EMB-8 (ortholog of human P450 oxidoreductase POR), and DAF-12 (homolog of human nuclear receptors VDR/LXR) constitute a hormone synthesis and nuclear receptor pathway in Caenorhabditis elegans This pathway specifically regulates the thermosensitive fusion of fat-storing lipid droplets. CYP-37A1, together with EMB-8, synthesizes a lipophilic hormone not identical to Δ7-dafachronic acid, which represses the fusion-promoting function of DAF-12. CYP-37A1 also negatively regulates thermotolerance and lifespan at high temperature in a DAF-12-dependent manner. Human CYP4V2 can substitute for CYP-37A1 in C. elegans This finding suggests the existence of a conserved CYP4V2-POR-nuclear receptor pathway that functions in converting multilocular lipid droplets to unilocular ones in human cells; misregulation of this pathway may lead to pathogenic fat storage.
Collapse
|
30
|
Praslicka B, Harmson JS, Kim J, Rangaraj VR, Ooi A, Gissendanner CR. BINDING SITE ANALYSIS OF THE CAENORHABDITIS ELEGANS NR4A NUCLEAR RECEPTOR NHR-6 DURING DEVELOPMENT. NUCLEAR RECEPTOR RESEARCH 2017; 4. [PMID: 29026837 DOI: 10.11131/2017/101288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Members of the NR4A subfamily of nuclear receptors make up a highly conserved, functionally diverse group of transcription factors implicated in a multitude of cellular processes such as proliferation, differentiation, apoptosis, metabolism and DNA repair. The gene nhr-6, which encodes the sole C. elegans NR4A nuclear receptor homolog, has a critical role in organogenesis and regulates the development of the spermatheca organ system. Our previous work revealed that nhr-6 is required for spermatheca cell divisions in late L3 and early L4 and spermatheca cell differentiation during the mid L4 stage. Here, we utilized chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq) to identify NHR-6 binding sites during both the late L3/early L4 and mid L4 developmental stages. Our results revealed 30,745 enriched binding sites for NHR-6, ~70% of which were within 3 kb upstream of a gene transcription start site. Binding sites for a cohort of candidate target genes with probable functions in spermatheca organogenesis were validated through qPCR. Reproductive and spermatheca phenotypes were also evaluated for these genes following a loss-of-function RNAi screen which revealed several genes with critical functions during spermatheca organogenesis. Our results uncovered a complex nuclear receptor regulatory network whereby NHR-6 regulates multiple cellular processes during spermatheca organogenesis.
Collapse
Affiliation(s)
- Brandon Praslicka
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, and Biology Program, School of Science, University of Louisiana Monroe, Monroe, LA, USA 71209
| | - Jeremy S Harmson
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, and Biology Program, School of Science, University of Louisiana Monroe, Monroe, LA, USA 71209
| | - Joohyun Kim
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA, USA 70803
| | - Vittobai Rashika Rangaraj
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, and Biology Program, School of Science, University of Louisiana Monroe, Monroe, LA, USA 71209
| | - Aikseng Ooi
- Department of Pharmacology & Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA 85721
| | - Chris R Gissendanner
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, and Biology Program, School of Science, University of Louisiana Monroe, Monroe, LA, USA 71209
| |
Collapse
|
31
|
Lažetić V, Fay DS. Molting in C. elegans. WORM 2017; 6:e1330246. [PMID: 28702275 DOI: 10.1080/21624054.2017.1330246] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
Molting is an essential developmental process for the majority of animal species on Earth. During the molting process, which is a specialized form of extracellular matrix (ECM) remodeling, the old apical ECM, or cuticle, is replaced with a new one. Many of the genes and pathways identified as important for molting in nematodes are highly conserved in vertebrates and include regulators and components of vesicular trafficking, steroid-hormone signaling, developmental timers, and hedgehog-like signaling. In this review, we discuss what is known about molting, with a focus on studies in Caenorhabditis elegans. We also describe the key structural elements of the cuticle that must be released, newly synthesized, or remodeled for proper molting to occur.
Collapse
Affiliation(s)
- Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, USA
| | - David S Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
32
|
Syed MH, Mark B, Doe CQ. Steroid hormone induction of temporal gene expression in Drosophila brain neuroblasts generates neuronal and glial diversity. eLife 2017; 6:26287. [PMID: 28394252 PMCID: PMC5403213 DOI: 10.7554/elife.26287] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/09/2017] [Indexed: 12/14/2022] Open
Abstract
An important question in neuroscience is how stem cells generate neuronal diversity. During Drosophila embryonic development, neural stem cells (neuroblasts) sequentially express transcription factors that generate neuronal diversity; regulation of the embryonic temporal transcription factor cascade is lineage-intrinsic. In contrast, larval neuroblasts generate longer ~50 division lineages, and currently only one mid-larval molecular transition is known: Chinmo/Imp/Lin-28+ neuroblasts transition to Syncrip+ neuroblasts. Here we show that the hormone ecdysone is required to down-regulate Chinmo/Imp and activate Syncrip, plus two late neuroblast factors, Broad and E93. We show that Seven-up triggers Chinmo/Imp to Syncrip/Broad/E93 transition by inducing expression of the Ecdysone receptor in mid-larval neuroblasts, rendering them competent to respond to the systemic hormone ecdysone. Importantly, late temporal gene expression is essential for proper neuronal and glial cell type specification. This is the first example of hormonal regulation of temporal factor expression in Drosophila embryonic or larval neural progenitors.
Collapse
Affiliation(s)
- Mubarak Hussain Syed
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Brandon Mark
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| |
Collapse
|
33
|
Nonaka S, Ando Y, Kanetani T, Hoshi C, Nakai Y, Nainu F, Nagaosa K, Shiratsuchi A, Nakanishi Y. Signaling pathway for phagocyte priming upon encounter with apoptotic cells. J Biol Chem 2017; 292:8059-8072. [PMID: 28325838 DOI: 10.1074/jbc.m116.769745] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/12/2017] [Indexed: 12/18/2022] Open
Abstract
The phagocytic elimination of cells undergoing apoptosis is an evolutionarily conserved innate immune mechanism for eliminating unnecessary cells. Previous studies showed an increase in the level of engulfment receptors in phagocytes after the phagocytosis of apoptotic cells, which leads to the enhancement of their phagocytic activity. However, precise mechanisms underlying this phenomenon require further clarification. We found that the pre-incubation of a Drosophila phagocyte cell line with the fragments of apoptotic cells enhanced the subsequent phagocytosis of apoptotic cells, accompanied by an augmented expression of the engulfment receptors Draper and integrin αPS3. The DNA-binding activity of the transcription repressor Tailless was transiently raised in those phagocytes, depending on two partially overlapping signal-transduction pathways for the induction of phagocytosis as well as the occurrence of engulfment. The RNAi knockdown of tailless in phagocytes abrogated the enhancement of both phagocytosis and engulfment receptor expression. Furthermore, the hemocyte-specific RNAi of tailless reduced apoptotic cell clearance in Drosophila embryos. Taken together, we propose the following mechanism for the activation of Drosophila phagocytes after an encounter with apoptotic cells: two partially overlapping signal-transduction pathways for phagocytosis are initiated; transcription repressor Tailless is activated; expression of engulfment receptors is stimulated; and phagocytic activity is enhanced. This phenomenon most likely ensures the phagocytic elimination of apoptotic cells by stimulated phagocytes and is thus considered as a mechanism to prime phagocytes in innate immunity.
Collapse
Affiliation(s)
- Saori Nonaka
- From the Graduate School of Medical Sciences and
| | - Yuki Ando
- From the Graduate School of Medical Sciences and
| | | | - Chiharu Hoshi
- School of Pharmacy, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yuji Nakai
- the Institute for Food Sciences, Hirosaki University, Aomori, Aomori 038-0012, Japan, and
| | - Firzan Nainu
- From the Graduate School of Medical Sciences and.,the Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi 90245, Indonesia
| | - Kaz Nagaosa
- the Institute for Food Sciences, Hirosaki University, Aomori, Aomori 038-0012, Japan, and
| | | | - Yoshinobu Nakanishi
- From the Graduate School of Medical Sciences and .,School of Pharmacy, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
34
|
Bodofsky S, Koitz F, Wightman B. CONSERVED AND EXAPTED FUNCTIONS OF NUCLEAR RECEPTORS IN ANIMAL DEVELOPMENT. NUCLEAR RECEPTOR RESEARCH 2017; 4:101305. [PMID: 29333434 PMCID: PMC5761748 DOI: 10.11131/2017/101305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The nuclear receptor gene family includes 18 members that are broadly conserved among multiple disparate animal phyla, indicating that they trace their evolutionary origins to the time at which animal life arose. Typical nuclear receptors contain two major domains: a DNA-binding domain and a C-terminal domain that may bind a lipophilic hormone. Many of these nuclear receptors play varied roles in animal development, including coordination of life cycle events and cellular differentiation. The well-studied genetic model systems of Drosophila, C. elegans, and mouse permit an evaluation of the extent to which nuclear receptor function in development is conserved or exapted (repurposed) over animal evolution. While there are some specific examples of conserved functions and pathways, there are many clear examples of exaptation. Overall, the evolutionary theme of exaptation appears to be favored over strict functional conservation. Despite strong conservation of DNA-binding domain sequences and activity, the nuclear receptors prove to be highly-flexible regulators of animal development.
Collapse
Affiliation(s)
- Shari Bodofsky
- Biology Department, Muhlenberg College, 2400 Chew St., Allentown, PA 18104
| | - Francine Koitz
- Biology Department, Muhlenberg College, 2400 Chew St., Allentown, PA 18104
| | - Bruce Wightman
- Biology Department, Muhlenberg College, 2400 Chew St., Allentown, PA 18104
| |
Collapse
|
35
|
Analysis of a lin-42/period Null Allele Implicates All Three Isoforms in Regulation of Caenorhabditis elegans Molting and Developmental Timing. G3-GENES GENOMES GENETICS 2016; 6:4077-4086. [PMID: 27729432 PMCID: PMC5144976 DOI: 10.1534/g3.116.034165] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Caenorhabditis elegans heterochronic gene pathway regulates the relative timing of events during postembryonic development. lin-42, the worm homolog of the circadian clock gene, period, is a critical element of this pathway. lin-42 function has been defined by a set of hypomorphic alleles that cause precocious phenotypes, in which later developmental events, such as the terminal differentiation of hypodermal cells, occur too early. A subset of alleles also reveals a significant role for lin-42 in molting; larval stages are lengthened and ecdysis often fails in these mutant animals. lin-42 is a complex locus, encoding overlapping and nonoverlapping isoforms. Although existing alleles that affect subsets of isoforms have illuminated important and distinct roles for this gene in developmental timing, molting, and the decision to enter the alternative dauer state, it is essential to have a null allele to understand all of the roles of lin-42 and its individual isoforms. To remedy this problem and discover the null phenotype, we engineered an allele that deletes the entire lin-42 protein-coding region. lin-42 null mutants are homozygously viable, but have more severe phenotypes than observed in previously characterized hypomorphic alleles. We also provide additional evidence for this conclusion by using the null allele as a base for reintroducing different isoforms, showing that each isoform can provide heterochronic and molting pathway activities. Transcript levels of the nonoverlapping isoforms appear to be under coordinate temporal regulation, despite being driven by independent promoters. The lin-42 null allele will continue to be an important tool for dissecting the functions of lin-42 in molting and developmental timing.
Collapse
|
36
|
Lu CJ, Tian BY, Cao Y, Zou CG, Zhang KQ. Nuclear receptor nhr-48 is required for pathogenicity of the second stage (J2) of the plant parasite Meloidogyne incognita. Sci Rep 2016; 6:34959. [PMID: 27762328 PMCID: PMC5071846 DOI: 10.1038/srep34959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/15/2016] [Indexed: 02/03/2023] Open
Abstract
Nuclear receptors (NRs) are a diverse class of transcription factors, which are involved in regulating a large number of physiological events in metazoans. However, the function of NRs is poorly understood in plant-parasitic nematodes. Here, members of the NR1J+K group of NRs in nematodes, including the free-living and plant parasites, were examined and phylogenetically analyzed. We found that the number of members of the NR1J+K group in plant-parasitic nematodes was less than that in the free-living nematodes, suggesting this reduction of NR1J+K group members in plant parasites maybe arose during the separation of the free-living and intermediately plant parasitic nematodes (Bursaphelenchus xylophilus). Interestingly, the DNA-binding domain (DBD) and ligand-binding domain (LBD) of NR1J+K members were separated into two gene locations in the plant parasites. Knockdown of Meloidogyne incognita WBMinc13296, the ortholog of Caenorhabditis elegans nhr-48 DBD, reduced infectivity, delayed development, and decreased reproductivity. J2 of M. incognita subjected to silencing of WBMinc13295, the orthologs of B. xylophilus nhr-48 LBD, exhibited developmental lag within the host and reduced reproductivity. This study provides new insights into the function of NRs and suggests that NRs are potential targets for developing effective strategies for biological control of plant-parasitic nematodes.
Collapse
Affiliation(s)
- Chao-Jun Lu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Bao-Yu Tian
- College of Life Science, Fujian Normal University, Fuzhou, Fujian 350108, China
| | - Yi Cao
- Key Laboratory of Molecular Genetics, Guizhou Academy of Tobacco Science, Guiyang, Guizhou 550081, China
| | - Cheng-Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Ke-Qin Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| |
Collapse
|
37
|
Melo CFOR, Esteves CZ, de Oliveira RN, Guerreiro TM, de Oliveira DN, Lima EDO, Miné JC, Allegretti SM, Catharino RR. Early developmental stages of Ascaris lumbricoides featured by high-resolution mass spectrometry. Parasitol Res 2016; 115:4107-4114. [PMID: 27412760 DOI: 10.1007/s00436-016-5183-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/23/2016] [Indexed: 11/26/2022]
Abstract
Ascaris lumbricoides is responsible for a highly disseminated helminth parasitic disease, ascariosis, a relevant parasitosis that responds for great financial burden on the public health system of developing countries. In this work, metabolic fingerprinting using high-resolution mass spectrometry (HRMS) was employed to identify marker molecules from A. lumbricoides in different development stages. We have identified nine biomarkers, such as pheromones and steroidal prohormones in early stages, among other molecules in late development stages, making up four molecules for fertilized eggs, four marker molecules for first larvae (L1) and one marker molecule for third larvae (L3). Therefore, our findings indicate that this approach is suitable for biochemical characterization of A. lumbricoides development stages. Moreover, the straightforward analytical method employed, with almost no sample preparation from a complex matrix (feces) using high-resolution mass spectrometry, suggests that it is possible to seek for an easier and faster way to study animal molding processes.
Collapse
Affiliation(s)
| | - Cibele Zanardi Esteves
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | - Tatiane Melina Guerreiro
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Diogo Noin de Oliveira
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Estela de Oliveira Lima
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Júlio César Miné
- Department of Clinical and Toxicological Analysis, State University of Ponta Grossa- UEPG, Ponta Grossa, Paraná, Brazil
| | | | - Rodrigo Ramos Catharino
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
38
|
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
|
39
|
Caratti G, Matthews L, Poolman T, Kershaw S, Baxter M, Ray D. Glucocorticoid receptor function in health and disease. Clin Endocrinol (Oxf) 2015; 83:441-8. [PMID: 25627931 DOI: 10.1111/cen.12728] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 12/17/2014] [Accepted: 01/19/2015] [Indexed: 12/20/2022]
Abstract
Glucocorticoid hormones are essential for life in vertebrates. They act through the glucocorticoid receptor (GR), which is expressed in virtually all cells of the human body. Yet the actions of glucocorticoids (GCs) are specific to particular cell types. Broadly GCs regulate carbohydrate metabolism, inflammation, stress and cell fate. Synthetic GCs are widely used in medicine and are by far the most frequent cause of Cushing's syndrome in routine practice. The advent of novel drugs targeting the GR offers new opportunities to treat patients with immune, or malignant disease, and may also offer new opportunities to manage patients with adrenal insufficiency also. This review covers the latest understanding of how GCs work, how their actions are affected by disease, and where the new drugs may take us.
Collapse
Affiliation(s)
- Giorgio Caratti
- Centre for Endocrinology and Diabetes, University of Manchester, Manchester, UK
| | - Laura Matthews
- Centre for Endocrinology and Diabetes, University of Manchester, Manchester, UK
| | - Toryn Poolman
- Centre for Endocrinology and Diabetes, University of Manchester, Manchester, UK
| | | | - Matthew Baxter
- Centre for Endocrinology and Diabetes, University of Manchester, Manchester, UK
| | - David Ray
- Centre for Endocrinology and Diabetes, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Hoffmann JM, Partridge L. Nuclear hormone receptors: Roles of xenobiotic detoxification and sterol homeostasis in healthy aging. Crit Rev Biochem Mol Biol 2015; 50:380-92. [PMID: 26383043 DOI: 10.3109/10409238.2015.1067186] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Health during aging can be improved by genetic, dietary and pharmacological interventions. Many of these increase resistance to various stressors, including xenobiotics. Up-regulation of xenobiotic detoxification genes is a transcriptomic signature shared by long-lived nematodes, flies and mice, suggesting that protection of cells from toxicity of xenobiotics may contribute to longevity. Expression of genes involved in xenobiotic detoxification is controlled by evolutionarily conserved transcriptional regulators. Three closely related subgroups of nuclear hormone receptors (NHRs) have a major role, and these include DAF-12 and NHR-8 in C. elegans, DHR96 in Drosophila and FXR, LXRs, PXR, CAR and VDR in mammals. In the invertebrates, these NHRs have been experimentally demonstrated to play a role in extension of lifespan by genetic and environmental interventions. NHRs represent critical hubs in that they regulate detoxification enzymes with broad substrate specificities, metabolizing both endo- and xeno-biotics. They also modulate homeostasis of steroid hormones and other endogenous cholesterol derivatives and lipid metabolism, and these roles, as well as xenobiotic detoxification, may contribute to the effects of NHRs on lifespan and health during aging, an issue that is being increasingly addressed in C. elegans and Drosophila. Disentangling the contribution of these processes to longevity will require more precise understanding of the molecular mechanisms by which each is effected, including identification of ligands and co-regulators of NHRs, patterns of tissue-specificity and mechanisms of interaction between tissues. The roles of vertebrate NHRs in determination of health during aging and lifespan have yet to be investigated.
Collapse
Affiliation(s)
| | - Linda Partridge
- a Max Planck Institute for Biology of Ageing , Cologne , Germany and.,b Institute of Healthy Ageing, and GEE (Genetics, Evolution and Environment), University College , London , UK
| |
Collapse
|
41
|
Mok DZL, Sternberg PW, Inoue T. Morphologically defined sub-stages of C. elegans vulval development in the fourth larval stage. BMC DEVELOPMENTAL BIOLOGY 2015; 15:26. [PMID: 26066484 PMCID: PMC4464634 DOI: 10.1186/s12861-015-0076-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/06/2023]
Abstract
Background During the fourth larval (L4) stage, vulval cells of C. elegans undergo extensive morphogenesis accompanied by changes in gene expression. This phase of vulval development, occurring after the well-studied induction of vulval cells, is not well understood but is potentially a useful context in which to study how a complex temporal sequence of events is regulated during development. However, a system for precisely describing different phases of vulval development in the L4 stage has been lacking. Results We defined ten sub-stages of L4 based on morphological criteria as observed using Nomarski microscopy (L4.0 ~ L4.9). Precise timing of each sub-stage at 20 °C was determined. We also re-examined the timing of expression for several gene expression markers, and correlated the sub-stages with the timing of other developmental events in the vulva and the uterus. Conclusions This scheme allows the developmental timing of an L4 individual to be determined at approximately one-hour resolution without the need to resort to time course experiments. These well-defined developmental stages will enable more precise description of gene expression and other developmental events.
Collapse
Affiliation(s)
- Darren Z L Mok
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore
| | - Paul W Sternberg
- HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Takao Inoue
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore. .,HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
42
|
Huang W, Xu F, Li J, Li L, Que H, Zhang G. Evolution of a novel nuclear receptor subfamily with emphasis on the member from the Pacific oyster Crassostrea gigas. Gene 2015; 567:164-72. [PMID: 25956376 DOI: 10.1016/j.gene.2015.04.082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/16/2015] [Accepted: 04/28/2015] [Indexed: 10/23/2022]
Abstract
Nuclear receptors (NRs) belong to the transcription factor superfamily that regulates development, homeostasis, differentiation, and reproduction in metazoans via control of gene expression. Recently, rapid advances in genome projects on various metazoans have provided new opportunities for studying the evolution and function of NRs. Typically structured NRs are divided into six subfamilies. Here, the gene for a typically structured NR (CgNR8A1) was cloned from the Pacific oyster Crassostrea gigas. However, this novel receptor could not be assigned to a known NR subfamily. By data mining, nine other CgNR8A1 gene homologs were identified in metazoans such as cnidarians, mollusks, annelids, echinoderms, hemichordates, and cephalochordates. Phylogenetic analysis showed that these receptors belonged to a novel NR subfamily, hereafter designated as NR8. Evolutionary analysis revealed that the NR8 subfamily was phylogenetically the third-oldest NR subfamily, and it originated from a common ancestor of Eumetazoa; several gene loss events occurred independently in ancestors of vertebrates, ecdysozoans, and platyhelminths, which do not have NR8 members. Furthermore, the function of CgNR8A1 was investigated to provide an insight into the functions of this novel NR subfamily. A nuclear localization signal peptide, GKHRNKKPRLD, was identified in CgNR8A1, and a recombinant full-length protein of CgNR8A1 was localized in the nuclei of HeLa cells. The mRNA expression profile of CgNR8A1 suggested that it might be involved in the embryogenesis of C. gigas. The electrophoretic mobility shift assay showed that CgNR8A1 binds strongly to conserved DNA core motifs DR0, DR2, and DR4 and weakly to DR1, DR3, DR5, Half, and Pal0. In summary, the novel NR8 subfamily identified in this study improves our understanding of NR evolution, and the functional analysis of CgNR8A1 provided further insights into the functions of NR8A1s.
Collapse
Affiliation(s)
- Wen Huang
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Xu
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Juan Li
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Li Li
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Huayong Que
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Guofan Zhang
- National & Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|
43
|
Praslicka B, Gissendanner CR. The C. elegans NR4A nuclear receptor gene nhr-6 promotes cell cycle progression in the spermatheca lineage. Dev Dyn 2015; 244:417-30. [PMID: 25529479 DOI: 10.1002/dvdy.24244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND NR4A nuclear receptors are a conserved, functionally diverse group of nuclear receptors that regulate multiple cellular processes including proliferation and differentiation. The gene nhr-6 encodes the sole Caenorhabditis elegans NR4A nuclear receptor homolog with an essential role in reproduction by regulating morphogenesis of the spermatheca, a somatic gonad organ involved in ovulation and fertilization. RESULTS Here, we identify the spermatheca cell lineage defects that occur in nhr-6 mutants. Utilizing cell marker analysis, we find that nhr-6 is required for cell cycle progression and that the cell proliferation phenotype is not due to premature cell cycle exit. We also show that loss of the negative cell cycle regulators fzr-1 and lin-35 suppresses the cell proliferation defects. We further demonstrate that NHR-6 activity intersects with Eph receptor signaling during spermatheca cell proliferation. CONCLUSIONS NHR-6 has an essential function in promoting cell cycle progression during G1 phase in a specific spermatheca cell lineage. Genetic suppression of the proliferation phenotype does not affect the differentiation phenotypes observed in nhr-6 mutants, indicating a dualistic role for nhr-6 in regulating cell proliferation and cell differentiation during spermatheca organogenesis.
Collapse
Affiliation(s)
- Brandon Praslicka
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | | |
Collapse
|
44
|
Hwang DS, Lee BY, Kim HS, Lee MC, Kyung DH, Om AS, Rhee JS, Lee JS. Genome-wide identification of nuclear receptor (NR) superfamily genes in the copepod Tigriopus japonicus. BMC Genomics 2014; 15:993. [PMID: 25407996 PMCID: PMC4247118 DOI: 10.1186/1471-2164-15-993] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 11/04/2014] [Indexed: 01/14/2023] Open
Abstract
Background Nuclear receptors (NRs) are a large superfamily of proteins defined by a DNA-binding domain (DBD) and a ligand-binding domain (LBD). They function as transcriptional regulators to control expression of genes involved in development, homeostasis, and metabolism. The number of NRs differs from species to species, because of gene duplications and/or lineage-specific gene losses during metazoan evolution. Many NRs in arthropods interact with the ecdysteroid hormone and are involved in ecdysone-mediated signaling in arthropods. The nuclear receptor superfamily complement has been reported in several arthropods, including crustaceans, but not in copepods. We identified the entire NR repertoire of the copepod Tigriopus japonicus, which is an important marine model species for ecotoxicology and environmental genomics. Results Using whole genome and transcriptome sequences, we identified a total of 31 nuclear receptors in the genome of T. japonicus. Nomenclature of the nuclear receptors was determined based on the sequence similarities of the DNA-binding domain (DBD) and ligand-binding domain (LBD). The 7 subfamilies of NRs separate into five major clades (subfamilies NR1, NR2, NR3, NR4, and NR5/6). Although the repertoire of NR members in, T. japonicus was similar to that reported for other arthropods, there was an expansion of the NR1 subfamily in Tigriopus japonicus. The twelve unique nuclear receptors identified in T. japonicus are members of NR1L. This expansion may be a unique lineage-specific feature of crustaceans. Interestingly, E78 and HR83, which are present in other arthropods, were absent from the genomes of T. japonicus and two congeneric copepod species (T. japonicus and Tigriopus californicus), suggesting copepod lineage-specific gene loss. Conclusions We identified all NR receptors present in the copepod, T. japonicus. Knowledge of the copepod nuclear receptor repertoire will contribute to a better understanding of copepod- and crustacean-specific NR evolution. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-993) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 440-746, South Korea.
| |
Collapse
|
45
|
Potential conservation of circadian clock proteins in the phylum Nematoda as revealed by bioinformatic searches. PLoS One 2014; 9:e112871. [PMID: 25396739 PMCID: PMC4232591 DOI: 10.1371/journal.pone.0112871] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/15/2014] [Indexed: 11/19/2022] Open
Abstract
Although several circadian rhythms have been described in C. elegans, its molecular clock remains elusive. In this work we employed a novel bioinformatic approach, applying probabilistic methodologies, to search for circadian clock proteins of several of the best studied circadian model organisms of different taxa (Mus musculus, Drosophila melanogaster, Neurospora crassa, Arabidopsis thaliana and Synechoccocus elongatus) in the proteomes of C. elegans and other members of the phylum Nematoda. With this approach we found that the Nematoda contain proteins most related to the core and accessory proteins of the insect and mammalian clocks, which provide new insights into the nematode clock and the evolution of the circadian system.
Collapse
|
46
|
Nelson MD, Trojanowski NF, George-Raizen JB, Smith CJ, Yu CC, Fang-Yen C, Raizen DM. The neuropeptide NLP-22 regulates a sleep-like state in Caenorhabditis elegans. Nat Commun 2014; 4:2846. [PMID: 24301180 PMCID: PMC3867200 DOI: 10.1038/ncomms3846] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/30/2013] [Indexed: 11/09/2022] Open
Abstract
Neuropeptides play central roles in the regulation of homeostatic behaviors such as sleep and feeding. Caenorhabditis elegans displays sleep-like quiescence of locomotion and feeding during a larval transition stage called lethargus and feeds during active larval and adult stages. Here we show that the neuropeptide NLP-22 is a regulator of Caenorhabditis elegans sleep-like quiescence observed during lethargus. nlp-22 shows cyclical mRNA expression in synchrony with lethargus; it is regulated by LIN-42, an orthologue of the core circadian protein PERIOD; and it is expressed solely in the two RIA interneurons. nlp-22 and the RIA interneurons are required for normal lethargus quiescence, and forced expression of nlp-22 during active stages causes anachronistic locomotion and feeding quiescence. Optogenetic stimulation of RIA interneurons has a movement-promoting effect, demonstrating functional complexity in a single neuron type. Our work defines a quiescence-regulating role for NLP-22 and expands our knowledge of the neural circuitry controlling Caenorhabditis elegans behavioral quiescence.
Collapse
Affiliation(s)
- M D Nelson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
A lophotrochozoan-specific nuclear hormone receptor is required for reproductive system development in the planarian. Dev Biol 2014; 396:150-7. [PMID: 25278423 DOI: 10.1016/j.ydbio.2014.09.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 11/24/2022]
Abstract
Germ cells of sexually reproducing organisms receive an array of cues from somatic tissues that instruct developmental processes. Although the nature of these signals differs amongst organisms, the importance of germline-soma interactions is a common theme. Recently, peptide hormones from the nervous system have been shown to regulate germ cell development in the planarian Schmidtea mediterranea; thus, we sought to investigate a second class of hormones with a conserved role in reproduction, the lipophilic hormones. In order to study these signals, we identified a set of putative lipophilic hormone receptors, known as nuclear hormone receptors, and analyzed their functions in reproductive development. We found one gene, nhr-1, belonging to a small class of functionally uncharacterized lophotrochozoan-specific receptors, to be essential for the development of differentiated germ cells. Upon nhr-1 knockdown, germ cells in the testes and ovaries fail to mature, and remain as undifferentiated germline stem cells. Further analysis revealed that nhr-1 mRNA is expressed in the accessory reproductive organs and is required for their development, suggesting that this transcription factor functions cell non-autonomously in regulating germ cell development. Our studies identify a role for nuclear hormone receptors in planarian reproductive maturation and reinforce the significance of germline-soma interactions in sexual reproduction across metazoans.
Collapse
|
48
|
Chow YL, Kawasaki Y, Sato F. Knockdown of the NHR-8 nuclear receptor enhanced sensitivity to the lipid-reducing activity of alkaloids in Caenorhabditis elegans. Biosci Biotechnol Biochem 2014; 78:2008-13. [PMID: 25052035 DOI: 10.1080/09168451.2014.940278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Caenorhabditis elegans is a versatile, whole-organism model for bioactivity screening. However, this worm has extensive defensive mechanisms against xenobiotics which limit its use for screening of pharmacologically active compounds. In this study, we report that knockdown of nhr-8, a gene involved in the xenobiotic response, increased the worm's sensitivity to the lipid-reducing effects of some isoquinoline alkaloids, especially berberine. On the other hand, crude extract of rhizome and cultured cells showed enhanced biological activity compared to the pure alkaloids in wild type worm, but this enhanced activity was not detected in nhr-8 RNAi worm, suggesting that some components in cell extracts might interfere with the defense response in this worm. The possibility of using C. elegans as a model for screening bioactive chemicals is discussed.
Collapse
Affiliation(s)
- Yit-Lai Chow
- a Department of Plant Gene and Totipotency , Graduate School of Biostudies, Kyoto University , Kyoto , Japan
| | | | | |
Collapse
|
49
|
Nuclear receptors in nematode development: Natural experiments made by a phylum. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:224-37. [PMID: 24984201 DOI: 10.1016/j.bbagrm.2014.06.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/21/2022]
Abstract
The development of complex multicellular organisms is dependent on regulatory decisions that are necessary for the establishment of specific differentiation and metabolic cellular states. Nuclear receptors (NRs) form a large family of transcription factors that play critical roles in the regulation of development and metabolism of Metazoa. Based on their DNA binding and ligand binding domains, NRs are divided into eight NR subfamilies from which representatives of six subfamilies are present in both deuterostomes and protostomes indicating their early evolutionary origin. In some nematode species, especially in Caenorhabditis, the family of NRs expanded to a large number of genes strikingly exceeding the number of NR genes in vertebrates or insects. Nematode NRs, including the multiplied Caenorhabditis genes, show clear relation to vertebrate and insect homologues belonging to six of the eight main NR subfamilies. This review summarizes advances in research of nematode NRs and their developmental functions. Nematode NRs can reveal evolutionarily conserved mechanisms that regulate specific developmental and metabolic processes as well as new regulatory adaptations. They represent the results of a large number of natural experiments with structural and functional potential of NRs for the evolution of the phylum. The conserved and divergent character of nematode NRs adds a new dimension to our understanding of the general biology of regulation by NRs. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
|
50
|
Vogeler S, Galloway TS, Lyons BP, Bean TP. The nuclear receptor gene family in the Pacific oyster, Crassostrea gigas, contains a novel subfamily group. BMC Genomics 2014; 15:369. [PMID: 24885009 PMCID: PMC4070562 DOI: 10.1186/1471-2164-15-369] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 04/30/2014] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Nuclear receptors are a superfamily of transcription factors important in key biological, developmental and reproductive processes. Several of these receptors are ligand- activated and through their ability to bind endogenous and exogenous ligands, are potentially vulnerable to xenobiotics. Molluscs are key ecological species in defining aquatic and terrestrial habitats and are sensitive to xenobiotic compounds in the environment. However, the understanding of nuclear receptor presence, function and xenobiotic disruption in the phylum Mollusca is limited. RESULTS Here, forty-three nuclear receptor sequences were mined from the genome of the Pacific oyster, Crassostrea gigas. They include members of NR0-NR5 subfamilies, notably lacking any NR6 members. Phylogenetic analyses of the oyster nuclear receptors have been conducted showing the presence of a large novel subfamily group not previously reported, which is named NR1P. Homologues to all previous identified nuclear receptors in other mollusc species have also been determined including the putative heterodimer partner retinoid X receptor, estrogen receptor and estrogen related receptor. CONCLUSION C. gigas contains a highly diverse set of nuclear receptors including a novel NR1 group, which provides important information on presence and evolution of this transcription factor superfamily in invertebrates. The Pacific oyster possesses two members of NR3, the sex steroid hormone receptor analogues, of which there are 9 in humans. This provides increasing evidence that steroid ligand specific expansion of this family is deuterostome specific. This new knowledge on divergence and emergence of nuclear receptors in C. gigas provides essential information for studying regulation of molluscan gene expression and the potential effects of xenobiotics.
Collapse
Affiliation(s)
- Susanne Vogeler
- />School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD UK
- />Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| | - Tamara S Galloway
- />School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD UK
| | - Brett P Lyons
- />Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| | - Tim P Bean
- />Centre for Environment, Fisheries and Aquaculture Science, Cefas Weymouth Laboratory, Barrack Road, Weymouth, DT4 8UB UK
| |
Collapse
|