1
|
Vora M, Dietz J, Wing Z, George K, Kelly Liu J, Rongo C, Savage-Dunn C. Genome-wide analysis of Smad and Schnurri transcription factors in C. elegans demonstrates widespread interaction and a function in collagen secretion. eLife 2025; 13:RP99394. [PMID: 39887187 PMCID: PMC11785376 DOI: 10.7554/elife.99394] [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] [Indexed: 02/01/2025] Open
Abstract
Smads and their transcription factor partners mediate the transcriptional responses of target cells to secreted ligands of the transforming growth factor-β (TGF-β) family, including those of the conserved bone morphogenetic protein (BMP) family, yet only a small number of direct target genes have been well characterized. In C. elegans, the BMP2/4 ortholog DBL-1 regulates multiple biological functions, including body size, via a canonical receptor-Smad signaling cascade. Here, we identify functional binding sites for SMA-3/Smad and its transcriptional partner SMA-9/Schnurri based on ChIP-seq peaks (identified by modEncode) and expression differences of nearby genes identified from RNA-seq analysis of corresponding mutants. We found that SMA-3 and SMA-9 have both overlapping and unique target genes. At a genome-wide scale, SMA-3/Smad acts as a transcriptional activator, whereas SMA-9/Schnurri direct targets include both activated and repressed genes. Mutations in sma-9 partially suppress the small body size phenotype of sma-3, suggesting some level of antagonism between these factors and challenging the prevailing model for Schnurri function. Functional analysis of target genes revealed a novel role in body size for genes involved in one-carbon metabolism and in the endoplasmic reticulum (ER) secretory pathway, including the disulfide reductase dpy-11. Our findings indicate that Smads and SMA-9/Schnurri have previously unappreciated complex genetic and genomic regulatory interactions that in turn regulate the secretion of extracellular components like collagen into the cuticle to mediate body size regulation.
Collapse
Affiliation(s)
- Mehul Vora
- Waksman Institute, Department of Genetics, Rutgers UniversityNew BrunswickUnited States
- ModOmics LtdSouthamptonUnited Kingdom
| | - Jonathan Dietz
- Waksman Institute, Department of Genetics, Rutgers UniversityNew BrunswickUnited States
| | - Zachary Wing
- Department of Biology, Queens College, CUNYNew YorkUnited States
| | - Karen George
- Waksman Institute, Department of Genetics, Rutgers UniversityNew BrunswickUnited States
| | - Jun Kelly Liu
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
| | - Christopher Rongo
- Waksman Institute, Department of Genetics, Rutgers UniversityNew BrunswickUnited States
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNYNew YorkUnited States
- PhD Program in Biology, The Graduate Center, CUNYNew YorkUnited States
| |
Collapse
|
2
|
Woodruff GC, Willis JH, Johnson E, Phillips PC. Widespread changes in gene expression accompany body size evolution in nematodes. G3 (BETHESDA, MD.) 2024; 14:jkae110. [PMID: 38775657 PMCID: PMC11304970 DOI: 10.1093/g3journal/jkae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 10/27/2023] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Body size is a fundamental trait that drives multiple evolutionary and ecological patterns. Caenorhabditis inopinata is a fig-associated nematode that is exceptionally large relative to other members of the genus, including Caenorhabditis elegans. We previously showed that C. inopinata is large primarily due to postembryonic cell size expansion that occurs during the larval-to-adult transition. Here, we describe gene expression patterns in C. elegans and C. inopinata throughout this developmental period to understand the transcriptional basis of body size change. We performed RNA-seq in both species across the L3, L4, and adult stages. Most genes are differentially expressed across all developmental stages, consistent with C. inopinata's divergent ecology and morphology. We also used a model comparison approach to identify orthologues with divergent dynamics across this developmental period between the 2 species. This included genes connected to neurons, behavior, stress response, developmental timing, and small RNA/chromatin regulation. Multiple hypodermal collagens were also observed to harbor divergent developmental dynamics across this period, and genes important for molting and body morphology were also detected. Genes associated with transforming growth factor β signaling revealed idiosyncratic and unexpected transcriptional patterns given their role in body size regulation in C. elegans. This widespread transcriptional divergence between these species is unexpected and maybe a signature of the ecological and morphological divergence of C. inopinata. Alternatively, transcriptional turnover may be the rule in the Caenorhabditis genus, indicative of widespread developmental system drift among species. This work lays the foundation for future functional genetic studies interrogating the bases of body size evolution in this group.
Collapse
Affiliation(s)
- Gavin C Woodruff
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
- School of Biological Sciences, University of Oklahoma, Norman, OK 73019, USA
| | - John H Willis
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Erik Johnson
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Patrick C Phillips
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
3
|
Hashizume O, Kawabe T, Funato Y, Miki H. Intestinal Mg 2+ accumulation induced by cnnm mutations decreases the body size by suppressing TORC2 signaling in Caenorhabditis elegans. Dev Biol 2024; 509:59-69. [PMID: 38373693 DOI: 10.1016/j.ydbio.2024.02.007] [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: 07/20/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Mg2+ is a vital ion involved in diverse cellular functions by forming complexes with ATP. Intracellular Mg2+ levels are tightly regulated by the coordinated actions of multiple Mg2+ transporters, such as the Mg2+ efflux transporter, cyclin M (CNNM). Caenorhabditis elegans (C. elegans) worms with mutations in both cnnm-1 and cnnm-3 exhibit excessive Mg2+ accumulation in intestinal cells, leading to various phenotypic abnormalities. In this study, we investigated the mechanism underlying the reduction in body size in cnnm-1; cnnm-3 mutant worms. RNA interference (RNAi) of gtl-1, which encodes a Mg2+-intake channel in intestinal cells, restored the worm body size, confirming that this phenotype is due to excessive Mg2+ accumulation. Moreover, RNAi experiments targeting body size-related genes and analyses of mutant worms revealed that the suppression of the target of rapamycin complex 2 (TORC2) signaling pathway was involved in body size reduction, resulting in downregulated DAF-7 expression in head ASI neurons. As the DAF-7 signaling pathway suppresses dauer formation under stress, cnnm-1; cnnm-3 mutant worms exhibited a greater tendency to form dauer upon induction. Collectively, our results revealed that excessive accumulation of Mg2+ repressed the TORC2 signaling pathway in C. elegans worms and suggest the novel role of the DAF-7 signaling pathway in the regulation of their body size.
Collapse
Affiliation(s)
- Osamu Hashizume
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan; Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tomofumi Kawabe
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yosuke Funato
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan; Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hiroaki Miki
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan; Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
4
|
Woodruff GC, Willis JH, Johnson E, Phillips PC. Widespread changes in gene expression accompany body size evolution in nematodes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564729. [PMID: 37961435 PMCID: PMC10635002 DOI: 10.1101/2023.10.30.564729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Body size is a fundamental trait that drives multiple evolutionary and ecological patterns. Caenorhabditis inopinata is a fig-associated nematode that is exceptionally large relative to other members of the genus, including C. elegans. We previously showed that C. inopinata is large primarily due to postembryonic cell size expansion that occurs during the larval-to-adult transition. Here, we describe gene expression patterns in C. elegans and C. inopinata throughout this developmental period to understand the transcriptional basis of body size change. We performed RNA-seq in both species across the L3, L4, and adult stages. Most genes are differentially expressed across all developmental stages, consistent with C. inopinata's divergent ecology and morphology. We also used a model comparison approach to identify orthologs with divergent dynamics across this developmental period between the two species. This included genes connected to neurons, behavior, stress response, developmental timing, and small RNA/chromatin regulation. Multiple hypodermal collagens were also observed to harbor divergent developmental dynamics across this period, and genes important for molting and body morphology were also detected. Genes associated with TGF-β signaling revealed idiosyncratic and unexpected transcriptional patterns given their role in body size regulation in C. elegans. Widespread transcriptional divergence between these species is unexpected and may be a signature of the ecological and morphological divergence of C. inopinata. Alternatively, transcriptional turnover may be the rule in the Caenorhabditis genus, indicative of widespread developmental system drift among species. This work lays the foundation for future functional genetic studies interrogating the bases of body size evolution in this group.
Collapse
Affiliation(s)
- Gavin C Woodruff
- University of Oregon, Eugene, Oregon, USA
- Current institution: University of Oklahoma, Norman, Oklahoma, USA
| | | | | | | |
Collapse
|
5
|
Zhang R, Fang J, Qi T, Zhu S, Yao L, Fang G, Li Y, Zang X, Xu W, Hao W, Liu S, Yang D, Chen D, Yang J, Ma X, Wu L. Maternal aging increases offspring adult body size via transmission of donut-shaped mitochondria. Cell Res 2023; 33:821-834. [PMID: 37500768 PMCID: PMC10624822 DOI: 10.1038/s41422-023-00854-8] [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: 12/09/2022] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Maternal age at childbearing has continued to increase in recent decades. However, whether and how it influences offspring adult traits are largely unknown. Here, using adult body size as the primary readout, we reveal that maternal rather than paternal age has an evolutionarily conserved effect on offspring adult traits in humans, Drosophila, and Caenorhabditis elegans. Elucidating the mechanisms of such effects in humans and other long-lived animals remains challenging due to their long life course and difficulties in conducting in vivo studies. We thus employ the short-lived and genetically tractable nematode C. elegans to explore the mechanisms underlying the regulation of offspring adult trait by maternal aging. By microscopic analysis, we find that old worms transmit aged mitochondria with a donut-like shape to offspring. These mitochondria are rejuvenated in the offspring's early life, with their morphology fully restored before adulthood in an AMPK-dependent manner. Mechanistically, we demonstrate that early-life mitochondrial dysfunction activates AMPK, which in turn not only alleviates mitochondrial abnormalities but also activates TGFβ signaling to increase offspring adult size. Together, our findings provide mechanistic insight into the ancient role of maternal aging in shaping the traits of adult offspring.
Collapse
Affiliation(s)
- Runshuai Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jinan Fang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Ting Qi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Shihao Zhu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Luxia Yao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Guicun Fang
- Microscopy Core Facility, Westlake University, Hangzhou, Zhejiang, China
| | - Yunsheng Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiao Zang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Weina Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shouye Liu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Dan Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Di Chen
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jian Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
DeGroot MS, Williams B, Chang TY, Maas Gamboa ML, Larus IM, Hong G, Fromme JC, Liu J. SMOC-1 interacts with both BMP and glypican to regulate BMP signaling in C. elegans. PLoS Biol 2023; 21:e3002272. [PMID: 37590248 PMCID: PMC10464977 DOI: 10.1371/journal.pbio.3002272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 08/29/2023] [Accepted: 07/22/2023] [Indexed: 08/19/2023] Open
Abstract
Secreted modular calcium-binding proteins (SMOCs) are conserved matricellular proteins found in organisms from Caenorhabditis elegans to humans. SMOC homologs characteristically contain 1 or 2 extracellular calcium-binding (EC) domain(s) and 1 or 2 thyroglobulin type-1 (TY) domain(s). SMOC proteins in Drosophila and Xenopus have been found to interact with cell surface heparan sulfate proteoglycans (HSPGs) to exert both positive and negative influences on the conserved bone morphogenetic protein (BMP) signaling pathway. In this study, we used a combination of biochemical, structural modeling, and molecular genetic approaches to dissect the functions of the sole SMOC protein in C. elegans. We showed that CeSMOC-1 binds to the heparin sulfate proteoglycan GPC3 homolog LON-2/glypican, as well as the mature domain of the BMP2/4 homolog DBL-1. Moreover, CeSMOC-1 can simultaneously bind LON-2/glypican and DBL-1/BMP. The interaction between CeSMOC-1 and LON-2/glypican is mediated specifically by the EC domain of CeSMOC-1, while the full interaction between CeSMOC-1 and DBL-1/BMP requires full-length CeSMOC-1. We provide both in vitro biochemical and in vivo functional evidence demonstrating that CeSMOC-1 functions both negatively in a LON-2/glypican-dependent manner and positively in a DBL-1/BMP-dependent manner to regulate BMP signaling. We further showed that in silico, Drosophila and vertebrate SMOC proteins can also bind to mature BMP dimers. Our work provides a mechanistic basis for how the evolutionarily conserved SMOC proteins regulate BMP signaling.
Collapse
Affiliation(s)
- Melisa S. DeGroot
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Byron Williams
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Timothy Y. Chang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Maria L. Maas Gamboa
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Isabel M. Larus
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Garam Hong
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - J. Christopher Fromme
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
7
|
DeGroot MS, Williams B, Chang TY, Maas Gamboa ML, Larus I, Fromme JC, Liu J. C. elegans SMOC-1 interacts with both BMP and glypican to regulate BMP signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.523017. [PMID: 36711863 PMCID: PMC9881921 DOI: 10.1101/2023.01.06.523017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Secreted modular calcium binding (SMOC) proteins are conserved matricellular proteins found in organisms from C. elegans to humans. SMOC homologs characteristically contain one or two extracellular calcium (EC) binding domain(s) and one or two thyroglobulin type-1 (TY) domain(s). SMOC proteins in Drosophila and Xenopus have been found to interact with cell surface heparan sulfate protein glycans (HSPGs) to exert both positive and negative influences on the conserved bone morphogenetic protein (BMP) signaling pathway. In this study, we used a combination of biochemical, structural modeling, and molecular genetic approaches to dissect the functions of the sole SMOC protein in C. elegans . We showed that SMOC-1 binds LON-2/glypican, as well as the mature domain of DBL-1/BMP. Moreover, SMOC-1 can simultaneously bind LON-2/glypican and DBL-1/BMP. The interaction between SMOC-1 and LON-2/glypican is mediated by the EC domain of SMOC-1, while the interaction between SMOC-1 and DBL-1/BMP involves full-length SMOC-1. We further showed that while SMOC-1(EC) is sufficient to promote BMP signaling when overexpressed, both the EC and TY domains are required for SMOC-1 function at the endogenous locus. Finally, when overexpressed, SMOC-1 can promote BMP signaling in the absence of LON-2/glypican. Taken together, our findings led to a model where SMOC-1 functions both negatively in a LON-2-dependent manner and positively in a LON-2-independent manner to regulate BMP signaling. Our work provides a mechanistic basis for how the evolutionarily conserved SMOC proteins regulate BMP signaling.
Collapse
Affiliation(s)
- Melisa S. DeGroot
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Byron Williams
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Timothy Y Chang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Maria L. Maas Gamboa
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Isabel Larus
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | | | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| |
Collapse
|
8
|
Arneaud SLB, McClendon J, Tatge L, Watterson A, Zuurbier KR, Madhu B, Gumienny TL, Douglas PM. Reduced bone morphogenic protein signaling along the gut-neuron axis by heat shock factor promotes longevity. Aging Cell 2022; 21:e13693. [PMID: 35977034 PMCID: PMC9470895 DOI: 10.1111/acel.13693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/24/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023] Open
Abstract
Aging is a complex and highly regulated process of interwoven signaling mechanisms. As an ancient transcriptional regulator of thermal adaptation and protein homeostasis, the Heat Shock Factor, HSF-1, has evolved functions within the nervous system to control age progression; however, the molecular details and signaling dynamics by which HSF-1 modulates age across tissues remain unclear. Herein, we report a nonautonomous mode of age regulation by HSF-1 in the Caenorhabditis elegans nervous system that works through the bone morphogenic protein, BMP, signaling pathway to modulate membrane trafficking in peripheral tissues. In particular, HSF-1 represses the expression of the neuron-specific BMP ligand, DBL-1, and initiates a complementary negative feedback loop within the intestine. By reducing receipt of DBL-1 in the periphery, the SMAD transcriptional coactivator, SMA-3, represses the expression of critical membrane trafficking regulators including Rab GTPases involved in early (RAB-5), late (RAB-7), and recycling (RAB-11.1) endosomal dynamics and the BMP receptor binding protein, SMA-10. This reduces cell surface residency and steady-state levels of the type I BMP receptor, SMA-6, in the intestine and further dampens signal transmission to the periphery. Thus, the ability of HSF-1 to coordinate BMP signaling along the gut-brain axis is an important determinate in age progression.
Collapse
Affiliation(s)
| | - Jacob McClendon
- Department of Molecular BiologyUT Southwestern Medical CenterDallasTexasUSA
| | - Lexus Tatge
- Department of Molecular BiologyUT Southwestern Medical CenterDallasTexasUSA
| | - Abigail Watterson
- Department of Molecular BiologyUT Southwestern Medical CenterDallasTexasUSA
| | - Kielen R. Zuurbier
- Department of Molecular BiologyUT Southwestern Medical CenterDallasTexasUSA
| | - Bhoomi Madhu
- Department of BiologyTexas Woman's UniversityDentonTexasUSA
| | | | - Peter M. Douglas
- Department of Molecular BiologyUT Southwestern Medical CenterDallasTexasUSA,Hamon Center for Regenerative Science and MedicineUT Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
9
|
Igreja C, Sommer RJ. The Role of Sulfation in Nematode Development and Phenotypic Plasticity. Front Mol Biosci 2022; 9:838148. [PMID: 35223994 PMCID: PMC8869759 DOI: 10.3389/fmolb.2022.838148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/25/2022] Open
Abstract
Sulfation is poorly understood in most invertebrates and a potential role of sulfation in the regulation of developmental and physiological processes of these organisms remains unclear. Also, animal model system approaches did not identify many sulfation-associated mechanisms, whereas phosphorylation and ubiquitination are regularly found in unbiased genetic and pharmacological studies. However, recent work in the two nematodes Caenorhabditis elegans and Pristionchus pacificus found a role of sulfatases and sulfotransferases in the regulation of development and phenotypic plasticity. Here, we summarize the current knowledge about the role of sulfation in nematodes and highlight future research opportunities made possible by the advanced experimental toolkit available in these organisms.
Collapse
Affiliation(s)
- Catia Igreja
- *Correspondence: Catia Igreja, ; Ralf J. Sommer,
| | | |
Collapse
|
10
|
Goodman MB, Savage-Dunn C. Reciprocal interactions between transforming growth factor beta signaling and collagens: Insights from Caenorhabditis elegans. Dev Dyn 2022; 251:47-60. [PMID: 34537996 PMCID: PMC8982858 DOI: 10.1002/dvdy.423] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
Studies in genetically tractable organisms such as the nematode Caenorhabditis elegans have led to pioneering insights into conserved developmental regulatory mechanisms. For example, Smad signal transducers for the transforming growth factor beta (TGF-β) superfamily were first identified in C. elegans and in the fruit fly Drosophila. Recent studies of TGF-β signaling and the extracellular matrix (ECM) in C. elegans have forged unexpected links between signaling and the ECM, yielding novel insights into the reciprocal interactions that occur across tissues and spatial scales, and potentially providing new opportunities for the study of biomechanical regulation of gene expression.
Collapse
Affiliation(s)
- Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University, CA 94304
| | - Cathy Savage-Dunn
- Department of Biology, Queens College at the City University of New York, 11367,Correspondence to: >
| |
Collapse
|
11
|
Clark JF, Ciccarelli EJ, Kayastha P, Ranepura G, Yamamoto KK, Hasan MS, Madaan U, Meléndez A, Savage-Dunn C. BMP pathway regulation of insulin signaling components promotes lipid storage in Caenorhabditis elegans. PLoS Genet 2021; 17:e1009836. [PMID: 34634043 PMCID: PMC8530300 DOI: 10.1371/journal.pgen.1009836] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/21/2021] [Accepted: 09/27/2021] [Indexed: 11/18/2022] Open
Abstract
A small number of peptide growth factor ligands are used repeatedly in development and homeostasis to drive programs of cell differentiation and function. Cells and tissues must integrate inputs from these diverse signals correctly, while failure to do so leads to pathology, reduced fitness, or death. Previous work using the nematode C. elegans identified an interaction between the bone morphogenetic protein (BMP) and insulin/IGF-1-like signaling (IIS) pathways in the regulation of lipid homeostasis. The molecular components required for this interaction, however, were not fully understood. Here we report that INS-4, one of 40 insulin-like peptides (ILPs), is regulated by BMP signaling to modulate fat accumulation. Furthermore, we find that the IIS transcription factor DAF-16/FoxO, but not SKN-1/Nrf, acts downstream of BMP signaling in lipid homeostasis. Interestingly, BMP activity alters sensitivity of these two transcription factors to IIS-promoted cytoplasmic retention in opposite ways. Finally, we probe the extent of BMP and IIS interactions by testing additional IIS functions including dauer formation, aging, and autophagy induction. Coupled with our previous work and that of other groups, we conclude that BMP and IIS pathways have at least three modes of interaction: independent, epistatic, and antagonistic. The molecular interactions we identify provide new insight into mechanisms of signaling crosstalk and potential therapeutic targets for IIS-related pathologies such as diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- James F. Clark
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Emma J. Ciccarelli
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Peter Kayastha
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Gehan Ranepura
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Katerina K. Yamamoto
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Muhammad S. Hasan
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Uday Madaan
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Alicia Meléndez
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Cathy Savage-Dunn
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
- * E-mail:
| |
Collapse
|
12
|
DeGroot MS, Greer R, Liu J. GPN-1/glypican and UNC-52/perlecan do not appear to function in BMP signaling to pattern the C. elegans postembryonic mesoderm. MICROPUBLICATION BIOLOGY 2021; 2021:10.17912/micropub.biology.000437. [PMID: 34405137 PMCID: PMC8363907 DOI: 10.17912/micropub.biology.000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/12/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) are diverse macromolecules consisting of a protein core modified with glycosaminoglycan (GAG) chains. HSPGs, including glypicans and perlecans, have been implicated in shaping the extracellular matrix (ECM) to affect growth factor signaling. Here, we tested if GPN-1/glypicanor UNC-52/perlecan plays a role in the bone morphogenetic protein (BMP) signaling pathway in patterning the C. elegans postembryonic mesoderm. Using the suppression of sma-9(0) (Susm)assay, we found that animals carrying mutant alleles of gpn-1 or unc-52 do not exhibit any Susm phenotype. We also tested and found that the two glypicans GPN-1 and LON-2 do not share functional redundancy in the BMP pathway. Our results suggest that GPN-1/glypican and UNC-52/perlecan do not play a major role in the C. elegans BMP pathway, at least in patterning of the postembryonic mesoderm.
Collapse
Affiliation(s)
- Melisa S DeGroot
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY. USA
| | - Robert Greer
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY. USA
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY. USA,
Correspondence to: Jun Liu ()
| |
Collapse
|
13
|
Hayes AJ, Melrose J. Neural Tissue Homeostasis and Repair Is Regulated via CS and DS Proteoglycan Motifs. Front Cell Dev Biol 2021; 9:696640. [PMID: 34409033 PMCID: PMC8365427 DOI: 10.3389/fcell.2021.696640] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
Chondroitin sulfate (CS) is the most abundant and widely distributed glycosaminoglycan (GAG) in the human body. As a component of proteoglycans (PGs) it has numerous roles in matrix stabilization and cellular regulation. This chapter highlights the roles of CS and CS-PGs in the central and peripheral nervous systems (CNS/PNS). CS has specific cell regulatory roles that control tissue function and homeostasis. The CNS/PNS contains a diverse range of CS-PGs which direct the development of embryonic neural axonal networks, and the responses of neural cell populations in mature tissues to traumatic injury. Following brain trauma and spinal cord injury, a stabilizing CS-PG-rich scar tissue is laid down at the defect site to protect neural tissues, which are amongst the softest tissues of the human body. Unfortunately, the CS concentrated in gliotic scars also inhibits neural outgrowth and functional recovery. CS has well known inhibitory properties over neural behavior, and animal models of CNS/PNS injury have demonstrated that selective degradation of CS using chondroitinase improves neuronal functional recovery. CS-PGs are present diffusely in the CNS but also form denser regions of extracellular matrix termed perineuronal nets which surround neurons. Hyaluronan is immobilized in hyalectan CS-PG aggregates in these perineural structures, which provide neural protection, synapse, and neural plasticity, and have roles in memory and cognitive learning. Despite the generally inhibitory cues delivered by CS-A and CS-C, some CS-PGs containing highly charged CS disaccharides (CS-D, CS-E) or dermatan sulfate (DS) disaccharides that promote neural outgrowth and functional recovery. CS/DS thus has varied cell regulatory properties and structural ECM supportive roles in the CNS/PNS depending on the glycoform present and its location in tissue niches and specific cellular contexts. Studies on the fruit fly, Drosophila melanogaster and the nematode Caenorhabditis elegans have provided insightful information on neural interconnectivity and the role of the ECM and its PGs in neural development and in tissue morphogenesis in a whole organism environment.
Collapse
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Wales, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and The Faculty of Medicine and Health, The University of Sydney, St. Leonard’s, NSW, Australia
| |
Collapse
|
14
|
Park A, Qiu Z, Lee M. Deletion of the RGD motif in LON-2/glypican is associated with morphological abnormalities. MICROPUBLICATION BIOLOGY 2021; 2021:10.17912/micropub.biology.000376. [PMID: 33718824 PMCID: PMC7948029 DOI: 10.17912/micropub.biology.000376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The lon-2 gene in Caenorhabditis elegans encodes a heparan sulfate proteoglycan family glypican that negatively regulates the BMP signaling pathway responsible for controlling body length. LON-2 contains multiple functional domains, including an RGD (Arg-Gly-Asp) motif at amino acid number from 348 to 350. A novel mutant allele of lon-2 was investigated in this study. In this mutant allele, lon-2(kq348ΔRGD), the RGD motif at position 348 was deleted. Another pre-existing mutant allele, lon-2(e678), contains a ~9kb deletion and lacks most of the genomic coding sequence. The lon-2(e678) line was used as a reference allele. The novel mutant line was significantly shorter than wild-type animals, suggesting that removal of the RGD motif in LON-2 may improve its ability to inhibit BMP signaling.
Collapse
Affiliation(s)
| | | | - Myeongwoo Lee
- Baylor University Department of Biology,
Correspondence to: Myeongwoo Lee ()
| |
Collapse
|
15
|
Lee S, Lim GE, Kim YN, Koo HS, Shim J. AP2M1 Supports TGF-β Signals to Promote Collagen Expression by Inhibiting Caveolin Expression. Int J Mol Sci 2021; 22:ijms22041639. [PMID: 33561975 PMCID: PMC7915421 DOI: 10.3390/ijms22041639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
The extracellular matrix (ECM) is important for normal development and disease states, including inflammation and fibrosis. To understand the complex regulation of ECM, we performed a suppressor screening using Caenorhabditis elegans expressing the mutant ROL-6 collagen protein. One cuticle mutant has a mutation in dpy-23 that encodes the μ2 adaptin (AP2M1) of clathrin-associated protein complex II (AP-2). The subsequent suppressor screening for dpy-23 revealed the lon-2 mutation. LON-2 functions to regulate body size through negative regulation of the tumor growth factor-beta (TGF-β) signaling pathway responsible for ECM production. RNA-seq analysis showed a dominant change in the expression of collagen genes and cuticle components. We noted an increase in the cav-1 gene encoding caveolin-1, which functions in clathrin-independent endocytosis. By knockdown of cav-1, the reduced TGF-β signal was significantly restored in the dpy-23 mutant. In conclusion, the dpy-23 mutation upregulated cav-1 expression in the hypodermis, and increased CAV-1 resulted in a decrease of TβRI. Finally, the reduction of collagen expression including rol-6 by the reduced TGF-β signal influenced the cuticle formation of the dpy-23 mutant. These findings could help us to understand the complex process of ECM regulation in organism development and disease conditions.
Collapse
Affiliation(s)
- Saerom Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si 10408, Gyeonggi-do, Korea; (S.L.); (G.-E.L.); (Y.-N.K.)
- Department of Biochemistry, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Ga-Eun Lim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si 10408, Gyeonggi-do, Korea; (S.L.); (G.-E.L.); (Y.-N.K.)
| | - Yong-Nyun Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si 10408, Gyeonggi-do, Korea; (S.L.); (G.-E.L.); (Y.-N.K.)
| | - Hyeon-Sook Koo
- Department of Biochemistry, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Correspondence: (H.-S.K.); (J.S.); Tel.: +82-2-2123-2695 (H.-S.K.); +82-31-920-2262 (J.S.)
| | - Jaegal Shim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si 10408, Gyeonggi-do, Korea; (S.L.); (G.-E.L.); (Y.-N.K.)
- Correspondence: (H.-S.K.); (J.S.); Tel.: +82-2-2123-2695 (H.-S.K.); +82-31-920-2262 (J.S.)
| |
Collapse
|
16
|
Tetraspanins TSP-12 and TSP-14 function redundantly to regulate the trafficking of the type II BMP receptor in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2020; 117:2968-2977. [PMID: 31988138 DOI: 10.1073/pnas.1918807117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tetraspanins are a unique family of 4-pass transmembrane proteins that play important roles in a variety of cell biological processes. We have previously shown that 2 paralogous tetraspanins in Caenorhabditis elegans, TSP-12 and TSP-14, function redundantly to promote bone morphogenetic protein (BMP) signaling. The underlying molecular mechanisms, however, are not fully understood. In this study, we examined the expression and subcellular localization patterns of endogenously tagged TSP-12 and TSP-14 proteins. We found that TSP-12 and TSP-14 share overlapping expression patterns in multiple cell types, and that both proteins are localized on the cell surface and in various types of endosomes, including early, late, and recycling endosomes. Animals lacking both TSP-12 and TSP-14 exhibit reduced cell-surface levels of the BMP type II receptor DAF-4/BMPRII, along with impaired endosome morphology and mislocalization of DAF-4/BMPRII to late endosomes and lysosomes. These findings indicate that TSP-12 and TSP-14 are required for the recycling of DAF-4/BMPRII. Together with previous findings that the type I receptor SMA-6 is recycled via the retromer complex, our work demonstrates the involvement of distinct recycling pathways for the type I and type II BMP receptors and highlights the importance of tetraspanin-mediated intracellular trafficking in the regulation of BMP signaling in vivo. As TSP-12 and TSP-14 are conserved in mammals, our findings suggest that the mammalian TSP-12 and TSP-14 homologs may also function in regulating transmembrane protein recycling and BMP signaling.
Collapse
|
17
|
Lakdawala MF, Madhu B, Faure L, Vora M, Padgett RW, Gumienny TL. Genetic interactions between the DBL-1/BMP-like pathway and dpy body size-associated genes in Caenorhabditis elegans. Mol Biol Cell 2019; 30:3151-3160. [PMID: 31693440 PMCID: PMC6938244 DOI: 10.1091/mbc.e19-09-0500] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/22/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling pathways control many developmental and homeostatic processes, including cell size and extracellular matrix remodeling. An understanding of how this pathway itself is controlled remains incomplete. To identify novel regulators of BMP signaling, we performed a forward genetic screen in Caenorhabditis elegans for genes involved in body size regulation, a trait under the control of BMP member DBL-1. We isolated mutations that suppress the long phenotype of lon-2, a gene that encodes a negative regulator that sequesters DBL-1. This screen was effective because we isolated alleles of several core components of the DBL-1 pathway, demonstrating the efficacy of the screen. We found additional alleles of previously identified but uncloned body size genes. Our screen also identified widespread involvement of extracellular matrix proteins in DBL-1 regulation of body size. We characterized interactions between the DBL-1 pathway and extracellular matrix and other genes that affect body morphology. We discovered that loss of some of these genes affects the DBL-1 pathway, and we provide evidence that DBL-1 signaling affects many molecular and cellular processes associated with body size. We propose a model in which multiple body size factors are controlled by signaling through the DBL-1 pathway and by DBL-1-independent processes.
Collapse
Affiliation(s)
| | - Bhoomi Madhu
- Department of Biology, Texas Woman’s University, Denton, TX 76204-5799
| | - Lionel Faure
- Department of Biology, Texas Woman’s University, Denton, TX 76204-5799
| | - Mehul Vora
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ 08854-8020
| | - Richard W. Padgett
- Waksman Institute of Microbiology, Rutgers University, Piscataway, NJ 08854-8020
- Waksman Institute of Microbiology Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854-8020
- Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08854-8020
| | - Tina L. Gumienny
- Department of Biology, Texas Woman’s University, Denton, TX 76204-5799
| |
Collapse
|
18
|
Sturm Á, Saskoi É, Tibor K, Weinhardt N, Vellai T. Highly efficient RNAi and Cas9-based auto-cloning systems for C. elegans research. Nucleic Acids Res 2019; 46:e105. [PMID: 29924347 PMCID: PMC6158509 DOI: 10.1093/nar/gky516] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Abstract
RNA interference (RNAi) technology used for the functional analysis of Caenorhabditis elegans genes frequently leads to phenotypes with low penetrance or even proves completely ineffective. The methods previously developed to solve this problem were built on mutant genetic backgrounds, such as those defective for rrf-3, in which endogenous RNAi pathways are overexpressed. These mutations, however, interferes with many other genetic pathways so that the detected phenotype cannot always be clearly linked to the RNAi-exposed gene. In addition, using RNAi-overexpressing mutant backgrounds requires time-consuming genetic crossing. Here, we present an improved RNAi vector that produces specific double-stranded RNA species only, and thereby significantly stronger phenotypes than the standard gene knockdown vector. The further advantage of the new RNAi vector is that the detected phenotype can be specifically linked to the gene silenced. We also created a new all-in-one C. elegans Cas9 vector whose spacer sequence is much easier to replace. Both new vectors include a novel CRISPR/Cas9-based auto-cloning vector system rendering needless the use of restriction and ligase enzymes in generating DNA constructs. This novel, efficient RNAi and auto-cloning Cas9 systems can be easily adapted to any other genetic model.
Collapse
Affiliation(s)
- Ádám Sturm
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Éva Saskoi
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Kovács Tibor
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Nóra Weinhardt
- Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
19
|
DeGroot MS, Shi H, Eastman A, McKillop AN, Liu J. The Caenorhabditis elegans SMOC-1 Protein Acts Cell Nonautonomously To Promote Bone Morphogenetic Protein Signaling. Genetics 2019; 211:683-702. [PMID: 30518528 PMCID: PMC6366928 DOI: 10.1534/genetics.118.301805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 12/04/2018] [Indexed: 01/20/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates many different developmental and homeostatic processes in metazoans. The BMP pathway is conserved in Caenorhabditis elegans, and is known to regulate body size and mesoderm development. We have identified the C. elegans smoc-1 (Secreted MOdular Calcium-binding protein-1) gene as a new player in the BMP pathway. smoc-1(0) mutants have a small body size, while overexpression of smoc-1 leads to a long body size and increased expression of the RAD-SMAD (reporter acting downstream of SMAD) BMP reporter, suggesting that SMOC-1 acts as a positive modulator of BMP signaling. Using double-mutant analysis, we showed that SMOC-1 antagonizes the function of the glypican LON-2 and acts through the BMP ligand DBL-1 to regulate BMP signaling. Moreover, SMOC-1 appears to specifically regulate BMP signaling without significant involvement in a TGFβ-like pathway that regulates dauer development. We found that smoc-1 is expressed in multiple tissues, including cells of the pharynx, intestine, and posterior hypodermis, and that the expression of smoc-1 in the intestine is positively regulated by BMP signaling. We further established that SMOC-1 functions cell nonautonomously to regulate body size. Human SMOC1 and SMOC2 can each partially rescue the smoc-1(0) mutant phenotype, suggesting that SMOC-1's function in modulating BMP signaling is evolutionarily conserved. Together, our findings highlight a conserved role of SMOC proteins in modulating BMP signaling in metazoans.
Collapse
Affiliation(s)
- Melisa S DeGroot
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Herong Shi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Alice Eastman
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Alexandra N McKillop
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| |
Collapse
|
20
|
Savage-Dunn C, Gleason RJ, Liu J, Padgett RW. Mutagenesis and Imaging Studies of BMP Signaling Mechanisms in C. elegans. Methods Mol Biol 2019; 1891:51-73. [PMID: 30414126 DOI: 10.1007/978-1-4939-8904-1_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
C. elegans has played a central role in the elucidation of the TGFβ pathway over the last two decades. This is due to the high conservation of the pathway components and the power of genetic and cell biological approaches applied toward understanding how the pathway signals. In Subheading 3, we detail approaches to study the BMP branch of the TGFβ pathway in C. elegans.
Collapse
Affiliation(s)
- Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY, Flushing, NY, USA
- PhD Programs in Biology and Biochemistry, The Graduate Center, CUNY, New York, NY, USA
| | - Ryan J Gleason
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Richard W Padgett
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ, USA.
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
21
|
Mazzochette EA, Nekimken AL, Loizeau F, Whitworth J, Huynh B, Goodman MB, Pruitt BL. The tactile receptive fields of freely moving Caenorhabditis elegans nematodes. Integr Biol (Camb) 2018; 10:450-463. [PMID: 30027970 PMCID: PMC6168290 DOI: 10.1039/c8ib00045j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Sensory neurons embedded in skin are responsible for the sense of touch. In humans and other mammals, touch sensation depends on thousands of diverse somatosensory neurons. By contrast, Caenorhabditis elegans nematodes have six gentle touch receptor neurons linked to simple behaviors. The classical touch assay uses an eyebrow hair to stimulate freely moving C. elegans, evoking evasive behavioral responses. This assay has led to the discovery of genes required for touch sensation, but does not provide control over stimulus strength or position. Here, we present an integrated system for performing automated, quantitative touch assays that circumvents these limitations and incorporates automated measurements of behavioral responses. The Highly Automated Worm Kicker (HAWK) unites a microfabricated silicon force sensor holding a glass bead forming the contact surface and video analysis with real-time force and position control. Using this system, we stimulated animals along the anterior-posterior axis and compared responses in wild-type and spc-1(dn) transgenic animals, which have a touch defect due to expression of a dominant-negative α-spectrin protein fragment. As expected from prior studies, delivering large stimuli anterior and posterior to the mid-point of the body evoked a reversal and a speed-up, respectively. The probability of evoking a response of either kind depended on stimulus strength and location; once initiated, the magnitude and quality of both reversal and speed-up behavioral responses were uncorrelated with stimulus location, strength, or the absence or presence of the spc-1(dn) transgene. Wild-type animals failed to respond when the stimulus was applied near the mid-point. These results show that stimulus strength and location govern the activation of a characteristic motor program and that the C. elegans body surface consists of two receptive fields separated by a gap.
Collapse
Affiliation(s)
- E A Mazzochette
- Department of Electrical Engineering, Stanford University, 94305, USA
| | - A L Nekimken
- Department of Mechanical Engineering, Stanford University, 94305, USA. and Department of Molecular and Cellular Physiology, Stanford University, 94305, USA
| | - F Loizeau
- Department of Mechanical Engineering, Stanford University, 94305, USA.
| | - J Whitworth
- Department of Mechanical Engineering, Stanford University, 94305, USA.
| | - B Huynh
- Department of Mechanical Engineering, Stanford University, 94305, USA.
| | - M B Goodman
- Department of Mechanical Engineering, Stanford University, 94305, USA. and Department of Molecular and Cellular Physiology, Stanford University, 94305, USA
| | - B L Pruitt
- Department of Mechanical Engineering, Stanford University, 94305, USA. and Department of Molecular and Cellular Physiology, Stanford University, 94305, USA and Department of Bioengineering, Stanford University, 94305, USA and Department of Mechanical Engineering, University of California, Santa Barbara, 93106, USA.
| |
Collapse
|
22
|
Philbrook A, Ramachandran S, Lambert CM, Oliver D, Florman J, Alkema MJ, Lemons M, Francis MM. Neurexin directs partner-specific synaptic connectivity in C. elegans. eLife 2018; 7:35692. [PMID: 30039797 PMCID: PMC6057746 DOI: 10.7554/elife.35692] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/21/2018] [Indexed: 01/14/2023] Open
Abstract
In neural circuits, individual neurons often make projections onto multiple postsynaptic partners. Here, we investigate molecular mechanisms by which these divergent connections are generated, using dyadic synapses in C. elegans as a model. We report that C. elegans nrx-1/neurexin directs divergent connectivity through differential actions at synapses with partnering neurons and muscles. We show that cholinergic outputs onto neurons are, unexpectedly, located at previously undefined spine-like protrusions from GABAergic dendrites. Both these spine-like features and cholinergic receptor clustering are strikingly disrupted in the absence of nrx-1. Excitatory transmission onto GABAergic neurons, but not neuromuscular transmission, is also disrupted. Our data indicate that NRX-1 located at presynaptic sites specifically directs postsynaptic development in GABAergic neurons. Our findings provide evidence that individual neurons can direct differential patterns of connectivity with their post-synaptic partners through partner-specific utilization of synaptic organizers, offering a novel view into molecular control of divergent connectivity. Nervous systems are complex networks of interconnected cells called neurons. These networks vary in size from a few hundred cells in worms, to tens of billions in the human brain. Within these networks, each individual neuron forms connections – called synapses – with many others. But these partner neurons are not necessarily alike. In fact, they may be different cell types. How neurons form distinct connections with different partner cells remains unclear. Part of the answer may lie in specialized proteins called cell adhesion molecules. These proteins occur on the cell surface and enable neurons to recognize one another. This helps ensure that the cells form appropriate connections via synapses. Cell adhesion molecules are therefore also known as synaptic organizers. Philbrook et al. have now examined the role of synaptic organizers in wiring up the nervous system of the nematode worm and model organism Caenorhabditis elegans. Motor neurons form connections with two types of partner cell: muscle cells and neurons. Philbrook et al. screened C. elegans that have mutations in genes encoding various synaptic organizers. This revealed that a protein called neurexin must be present for motor neurons to form synapses with other neurons. By contrast, neurexin is not required for the same neurons to establish synapses with muscles. Philbrook et al. found that neuron-to-neuron synapses arise at specialized finger-like projections. These resemble the dendritic spines at which synapses form in the brains of mammals, and had not been previously identified in C. elegans. In worms that lack neurexin, these spine-like structures do not form correctly, disrupting the formation of neuron-to-neuron connections. Previous work has implicated neurexin in synapse formation in the mammalian brain. But this is the first study to reveal a role for neurexin in establishing partner-specific synaptic connections. Mutations in synaptic organizers, including neurexin, contribute to disorders of brain development. These include schizophrenia and autism spectrum disorders. Learning more about how neurexin helps establish specific synaptic connections may help us understand how these disorders arise.
Collapse
Affiliation(s)
- Alison Philbrook
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Shankar Ramachandran
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Christopher M Lambert
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Devyn Oliver
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Jeremy Florman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Mark J Alkema
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Michele Lemons
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States.,Department of Natural Sciences, Assumption College, Worcester, United States
| | - Michael M Francis
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
23
|
The Heterochronic Gene lin-14 Controls Axonal Degeneration in C. elegans Neurons. Cell Rep 2018; 20:2955-2965. [PMID: 28930688 DOI: 10.1016/j.celrep.2017.08.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/31/2017] [Accepted: 08/25/2017] [Indexed: 01/23/2023] Open
Abstract
The disproportionate length of an axon makes its structural and functional maintenance a major task for a neuron. The heterochronic gene lin-14 has previously been implicated in regulating the timing of key developmental events in the nematode C. elegans. Here, we report that LIN-14 is critical for maintaining neuronal integrity. Animals lacking lin-14 display axonal degeneration and guidance errors in both sensory and motor neurons. We demonstrate that LIN-14 functions both cell autonomously within the neuron and non-cell autonomously in the surrounding tissue, and we show that interaction between the axon and its surrounding tissue is essential for the preservation of axonal structure. Furthermore, we demonstrate that lin-14 expression is only required during a short period early in development in order to promote axonal maintenance throughout the animal's life. Our results identify a crucial role for LIN-14 in preventing axonal degeneration and in maintaining correct interaction between an axon and its surrounding tissue.
Collapse
|
24
|
Clark JF, Meade M, Ranepura G, Hall DH, Savage-Dunn C. Caenorhabditis elegans DBL-1/BMP Regulates Lipid Accumulation via Interaction with Insulin Signaling. G3 (BETHESDA, MD.) 2018; 8:343-351. [PMID: 29162682 PMCID: PMC5765361 DOI: 10.1534/g3.117.300416] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/18/2017] [Indexed: 11/23/2022]
Abstract
Metabolic homeostasis is coordinately controlled by diverse inputs. Understanding these regulatory networks is vital to combating metabolic disorders. The nematode Caenorhabditis elegans has emerged as a powerful, genetically tractable model system for the discovery of lipid regulatory mechanisms. Here we introduce DBL-1, the C. elegans homolog of bone morphogenetic protein 2/4 (BMP2/4), as a significant regulator of lipid homeostasis. We used neutral lipid staining and a lipid droplet marker to demonstrate that both increases and decreases in DBL-1/BMP signaling result in reduced lipid stores and lipid droplet count. We find that lipid droplet size, however, correlates positively with the level of DBL-1/BMP signaling. Regulation of lipid accumulation in the intestine occurs through non-cell-autonomous signaling, since expression of SMA-3, a Smad signal transducer, in the epidermis (hypodermis) is sufficient to rescue the loss of lipid accumulation. Finally, genetic evidence indicates that DBL-1/BMP functions upstream of Insulin/IGF-1 Signaling in lipid metabolism. We conclude that BMP signaling regulates lipid metabolism in C. elegans through interorgan signaling to the Insulin pathway, shedding light on a less well-studied regulatory mechanism for metabolic homeostasis.
Collapse
Affiliation(s)
- James F Clark
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York, New York 10016
- Biology Department, Queens College, CUNY, Flushing, New York 11367
| | - Michael Meade
- Biology Department, Queens College, CUNY, Flushing, New York 11367
| | - Gehan Ranepura
- Biology Department, Queens College, CUNY, Flushing, New York 11367
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Cathy Savage-Dunn
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York, New York 10016
- Biology Department, Queens College, CUNY, Flushing, New York 11367
| |
Collapse
|
25
|
Neural Glycosylphosphatidylinositol-Anchored Proteins in Synaptic Specification. Trends Cell Biol 2017; 27:931-945. [PMID: 28743494 DOI: 10.1016/j.tcb.2017.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins are a specialized class of lipid-associated neuronal membrane proteins that perform diverse functions in the dynamic control of axon guidance, synaptic adhesion, cytoskeletal remodeling, and localized signal transduction, particularly at lipid raft domains. Recent studies have demonstrated that a subset of GPI-anchored proteins act as critical regulators of synapse development by modulating specific synaptic adhesion pathways via direct interactions with key synapse-organizing proteins. Additional studies have revealed that alteration of these regulatory mechanisms may underlie various brain disorders. In this review, we highlight the emerging role of GPI-anchored proteins as key synapse organizers that aid in shaping the properties of various types of synapses and circuits in mammals.
Collapse
|
26
|
Abstract
Transforming growth factor β (TGF-β) and related ligands have potent effects on an enormous diversity of biological functions in all animals examined. Because of the strong conservation of TGF-β family ligand functions and signaling mechanisms, studies from multiple animal systems have yielded complementary and synergistic insights. In the nematode Caenorhabditis elegans, early studies were instrumental in the elucidation of TGF-β family signaling mechanisms. Current studies in C. elegans continue to identify new functions for the TGF-β family in this organism as well as new conserved mechanisms of regulation.
Collapse
Affiliation(s)
- Cathy Savage-Dunn
- Department of Biology, Queens College, and the Graduate Center, New York, New York 11367
| | - Richard W Padgett
- Waksman Institute, Department of Molecular Biology and Biochemistry, Cancer Institute of New Jersey, Rutgers University, Piscataway, New Jersey 08854-8020
| |
Collapse
|
27
|
Sharma V, Roy S, Sekler I, O'Halloran DM. The NCLX-type Na +/Ca 2+ Exchanger NCX-9 Is Required for Patterning of Neural Circuits in Caenorhabditis elegans. J Biol Chem 2017; 292:5364-5377. [PMID: 28196860 DOI: 10.1074/jbc.m116.758953] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 02/13/2017] [Indexed: 12/22/2022] Open
Abstract
NCLX is a Na+/Ca2+ exchanger that uses energy stored in the transmembrane sodium gradient to facilitate the exchange of sodium ions for ionic calcium. Mammals have a single NCLX, which has been shown to function primarily at the mitochondrion and is an important regulator of neuronal physiology by contributing to neurotransmission and synaptic plasticity. The role of NCLX in developmental cell patterning (e.g. in neural circuits) is largely unknown. Here we describe a novel role for the Caenorhabditis elegans NCLX-type protein, NCX-9, in neural circuit formation. NCX-9 functions in hypodermal seam cells that secrete the axon guidance cue UNC-129/BMP, and our data revealed that ncx-9-/- mutant animals exhibit development defects in stereotyped left/right axon guidance choices within the GABAergic motor neuron circuit. Our data also implicate NCX-9 in a LON-2/heparan sulfate and UNC-6/netrin-mediated, RAC-dependent signaling pathway to guide left/right patterning within this circuit. Finally, we also provide in vitro physiology data supporting the role for NCX-9 in handling calcium exchange at the mitochondrion. Taken together, our work reveals the specificity by which the handling by NCLX of calcium exchange can map to neural circuit patterning and axon guidance decisions during development.
Collapse
Affiliation(s)
- Vishal Sharma
- From the Department of Biological Sciences and.,the Institute for Neuroscience, George Washington University, Washington, D. C. 20052 and
| | - Soumitra Roy
- the Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8499000, Israel
| | - Israel Sekler
- the Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8499000, Israel
| | - Damien M O'Halloran
- From the Department of Biological Sciences and .,the Institute for Neuroscience, George Washington University, Washington, D. C. 20052 and
| |
Collapse
|
28
|
Fukunaga T, Iwasaki W. Inactivity periods and postural change speed can explain atypical postural change patterns of Caenorhabditis elegans mutants. BMC Bioinformatics 2017; 18:46. [PMID: 28103804 PMCID: PMC5244558 DOI: 10.1186/s12859-016-1408-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/06/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With rapid advances in genome sequencing and editing technologies, systematic and quantitative analysis of animal behavior is expected to be another key to facilitating data-driven behavioral genetics. The nematode Caenorhabditis elegans is a model organism in this field. Several video-tracking systems are available for automatically recording behavioral data for the nematode, but computational methods for analyzing these data are still under development. RESULTS In this study, we applied the Gaussian mixture model-based binning method to time-series postural data for 322 C. elegans strains. We revealed that the occurrence patterns of the postural states and the transition patterns among these states have a relationship as expected, and such a relationship must be taken into account to identify strains with atypical behaviors that are different from those of wild type. Based on this observation, we identified several strains that exhibit atypical transition patterns that cannot be fully explained by their occurrence patterns of postural states. Surprisingly, we found that two simple factors-overall acceleration of postural movement and elimination of inactivity periods-explained the behavioral characteristics of strains with very atypical transition patterns; therefore, computational analysis of animal behavior must be accompanied by evaluation of the effects of these simple factors. Finally, we found that the npr-1 and npr-3 mutants have similar behavioral patterns that were not predictable by sequence homology, proving that our data-driven approach can reveal the functions of genes that have not yet been characterized. CONCLUSION We propose that elimination of inactivity periods and overall acceleration of postural change speed can explain behavioral phenotypes of strains with very atypical postural transition patterns. Our methods and results constitute guidelines for effectively finding strains that show "truly" interesting behaviors and systematically uncovering novel gene functions by bioimage-informatic approaches.
Collapse
Affiliation(s)
- Tsukasa Fukunaga
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8568, Japan. .,Faculty of Science and Engineering, Waseda University, Tokyo, 169-0072, Japan. .,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan.
| | - Wataru Iwasaki
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8568, Japan. .,Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, 277-8564, Japan. .,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0032, Japan.
| |
Collapse
|
29
|
Wang L, Liu Z, Shi H, Liu J. Two Paralogous Tetraspanins TSP-12 and TSP-14 Function with the ADAM10 Metalloprotease SUP-17 to Promote BMP Signaling in Caenorhabditis elegans. PLoS Genet 2017; 13:e1006568. [PMID: 28068334 PMCID: PMC5261805 DOI: 10.1371/journal.pgen.1006568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/24/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
The highly conserved bone morphogenetic protein (BMP) signaling pathway regulates many developmental and homeostatic processes. While the core components of the BMP pathway have been well studied, much research is needed for understanding the mechanisms involved in the precise spatiotemporal control of BMP signaling in vivo. Here, we provide evidence that two paralogous and evolutionarily conserved tetraspanins, TSP-12 and TSP-14, function redundantly to promote BMP signaling in C. elegans. We further show that the ADAM10 (adisintegrin and metalloprotease 10) ortholog SUP-17 also functions to promote BMP signaling, and that TSP-12 can bind to and promote the cell surface localization of SUP-17. SUP-17/ADAM10 is known to be involved in the ligand-induced proteolytic processing of the Notch receptor. We have evidence that the function of SUP-17, and of TSP-12/TSP-14 in BMP signaling is independent of their roles in Notch signaling. Furthermore, presenilins, core components of the γ-secretase complex involved in processing Notch, do not appear to play a role in BMP signaling. These studies established a new role of the TSP-12/TSP-14/SUP-17 axis in regulating BMP signaling, in addition to their known function in the Notch signaling pathway. We also provide genetic evidence showing that a known BMP signaling modulator, UNC-40/neogenin/DCC, is one of the substrates of SUP-17/ADAM10 in the BMP signaling pathway. Bone morphogenetic protein (BMP) signaling regulates multiple developmental and homeostatic processes. Misregulation of this pathway can cause various diseases, including cancers. Thus, it is essential to understand how BMP signaling is tightly regulated spatiotemporally in vivo. We have identified a highly conserved ADAM (a disintegrin and metalloprotease) protein, SUP-17/ADAM10, as an important factor in modulating BMP signaling in C. elegans. We showed that the proper localization and function of this ADAM protease require two conserved tetraspanin proteins, TSP-12 and TSP-14. We provided genetic evidence showing that one of the substrates of SUP-17/ADAM10 in the BMP signaling pathway is a known BMP signaling modulator, UNC-40/neogenin/DCC. Our studies established a new role of the TSP-12-TSP-14-SUP-17 axis in regulating BMP signaling, in addition to and independent of their known function in the Notch signaling pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Zhiyu Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Herong Shi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
30
|
Blanchette CR, Thackeray A, Perrat PN, Hekimi S, Bénard CY. Functional Requirements for Heparan Sulfate Biosynthesis in Morphogenesis and Nervous System Development in C. elegans. PLoS Genet 2017; 13:e1006525. [PMID: 28068429 PMCID: PMC5221758 DOI: 10.1371/journal.pgen.1006525] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/06/2016] [Indexed: 12/28/2022] Open
Abstract
The regulation of cell migration is essential to animal development and physiology. Heparan sulfate proteoglycans shape the interactions of morphogens and guidance cues with their respective receptors to elicit appropriate cellular responses. Heparan sulfate proteoglycans consist of a protein core with attached heparan sulfate glycosaminoglycan chains, which are synthesized by glycosyltransferases of the exostosin (EXT) family. Abnormal HS chain synthesis results in pleiotropic consequences, including abnormal development and tumor formation. In humans, mutations in either of the exostosin genes EXT1 and EXT2 lead to osteosarcomas or multiple exostoses. Complete loss of any of the exostosin glycosyltransferases in mouse, fish, flies and worms leads to drastic morphogenetic defects and embryonic lethality. Here we identify and study previously unavailable viable hypomorphic mutations in the two C. elegans exostosin glycosyltransferases genes, rib-1 and rib-2. These partial loss-of-function mutations lead to a severe reduction of HS levels and result in profound but specific developmental defects, including abnormal cell and axonal migrations. We find that the expression pattern of the HS copolymerase is dynamic during embryonic and larval morphogenesis, and is sustained throughout life in specific cell types, consistent with HSPGs playing both developmental and post-developmental roles. Cell-type specific expression of the HS copolymerase shows that HS elongation is required in both the migrating neuron and neighboring cells to coordinate migration guidance. Our findings provide insights into general principles underlying HSPG function in development. During animal development, cells and neurons navigate long distances to reach their final target destinations. Migrating cells are guided by extracellular molecular cues, and cellular responses to these cues are regulated by heparan sulfate proteoglycans. Heparan sulfate proteoglycans are proteins with long heparan sulfate polysaccharide chains attached. Here we identify and study previously unavailable viable mutants that disrupt the elongation of the heparan sulfate chains in the nematode C. elegans. Our analysis shows that these HS-chain-elongation mutations affect the development of the nervous system as they result in misguided migrations of neurons and axons. Furthermore, we find that heparan sulfate chain elongation occurs in numerous cell types during development and that the coordinated production of heparan sulfate proteoglycans, in both the migrating cell and neighboring tissues, ensures proper migration. Our findings highlight the critical roles of heparan sulfate proteoglycans in nervous system development and the evolutionary conservation of the molecular mechanisms driving guided migrations.
Collapse
Affiliation(s)
- Cassandra R. Blanchette
- Department of Neurobiology, UMass Medical School, Worcester, Massachusetts, United States of America
| | - Andrea Thackeray
- Department of Neurobiology, UMass Medical School, Worcester, Massachusetts, United States of America
| | - Paola N. Perrat
- Department of Neurobiology, UMass Medical School, Worcester, Massachusetts, United States of America
| | | | - Claire Y. Bénard
- Department of Neurobiology, UMass Medical School, Worcester, Massachusetts, United States of America
- Department of Biological Sciences, University of Quebec at Montreal, Montreal, Canada
- * E-mail: ,
| |
Collapse
|
31
|
Abstract
The discovery of the transforming growth factor β (TGF-β) family ligands and the realization that their bioactivities need to be tightly controlled temporally and spatially led to intensive research that has identified a multitude of extracellular modulators of TGF-β family ligands, uncovered their functions in developmental and pathophysiological processes, defined the mechanisms of their activities, and explored potential modulator-based therapeutic applications in treating human diseases. These studies revealed a diverse repertoire of extracellular and membrane-associated molecules that are capable of modulating TGF-β family signals via control of ligand availability, processing, ligand-receptor interaction, and receptor activation. These molecules include not only soluble ligand-binding proteins that were conventionally considered as agonists and antagonists of TGF-β family of growth factors, but also extracellular matrix (ECM) proteins and proteoglycans that can serve as "sink" and control storage and release of both the TGF-β family ligands and their regulators. This extensive network of soluble and ECM modulators helps to ensure dynamic and cell-specific control of TGF-β family signals. This article reviews our knowledge of extracellular modulation of TGF-β growth factors by diverse proteins and their molecular mechanisms to regulate TGF-β family signaling.
Collapse
Affiliation(s)
- Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
32
|
The Caenorhabditis elegans Ephrin EFN-4 Functions Non-cell Autonomously with Heparan Sulfate Proteoglycans to Promote Axon Outgrowth and Branching. Genetics 2015; 202:639-60. [PMID: 26645816 DOI: 10.1534/genetics.115.185298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/02/2015] [Indexed: 01/21/2023] Open
Abstract
The Eph receptors and their cognate ephrin ligands play key roles in many aspects of nervous system development. These interactions typically occur within an individual tissue type, serving either to guide axons to their terminal targets or to define boundaries between the rhombomeres of the hindbrain. We have identified a novel role for the Caenorhabditis elegans ephrin EFN-4 in promoting primary neurite outgrowth in AIY interneurons and D-class motor neurons. Rescue experiments reveal that EFN-4 functions non-cell autonomously in the epidermis to promote primary neurite outgrowth. We also find that EFN-4 plays a role in promoting ectopic axon branching in a C. elegans model of X-linked Kallmann syndrome. In this context, EFN-4 functions non-cell autonomously in the body-wall muscle and in parallel with HS modification genes and HSPG core proteins. This is the first report of an epidermal ephrin providing a developmental cue to the nervous system.
Collapse
|
33
|
Klammert U, Mueller TD, Hellmann TV, Wuerzler KK, Kotzsch A, Schliermann A, Schmitz W, Kuebler AC, Sebald W, Nickel J. GDF-5 can act as a context-dependent BMP-2 antagonist. BMC Biol 2015; 13:77. [PMID: 26385096 PMCID: PMC4575486 DOI: 10.1186/s12915-015-0183-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/27/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein (BMP)-2 and growth and differentiation factor (GDF)-5 are two related transforming growth factor (TGF)-β family members with important functions in embryonic development and tissue homeostasis. BMP-2 is best known for its osteoinductive properties whereas GDF-5-as evident from its alternative name, cartilage derived morphogenetic protein 1-plays an important role in the formation of cartilage. In spite of these differences both factors signal by binding to the same subset of BMP receptors, raising the question how these different functionalities are generated. The largest difference in receptor binding is observed in the interaction with the type I receptor BMPR-IA. GDF-5, in contrast to BMP-2, shows preferential binding to the isoform BMPR-IB, which is abrogated by a single amino acid (A57R) substitution. The resulting variant, GDF-5 R57A, represents a "BMP-2 mimic" with respect to BMP receptor binding. In this study we thus wanted to analyze whether the two growth factors can induce distinct signals via an identically composed receptor. RESULTS Unexpectedly and dependent on the cellular context, GDF-5 R57A showed clear differences in its activity compared to BMP-2. In ATDC-5 cells, both ligands induced alkaline phosphatase (ALP) expression with similar potency. But in C2C12 cells, the BMP-2 mimic GDF-5 R57A (and also wild-type GDF-5) clearly antagonized BMP-2-mediated ALP expression, despite signaling in both cell lines occurring solely via BMPR-IA. The BMP-2- antagonizing properties of GDF-5 and GDF-5 R57A could also be observed in vivo when implanting BMP-2 and either one of the two GDF-5 ligands simultaneously at heterotopic sites. CONCLUSIONS Although comparison of the crystal structures of the GDF-5 R57A:BMPR-IAEC- and BMP-2:BMPR-IAEC complex revealed small ligand-specific differences, these cannot account for the different signaling characteristics because the complexes seem identical in both differently reacting cell lines. We thus predict an additional component, most likely a not yet identified GDF-5-specific co-receptor, which alters the output of the signaling complexes. Hence the presence or absence of this component then switches GDF-5's signaling capabilities to act either similar to BMP-2 or as a BMP-2 antagonist. These findings might shed new light on the role of GDF-5, e.g., in cartilage maintenance and/or limb development in that it might act as an inhibitor of signaling events initiated by other BMPs.
Collapse
Affiliation(s)
- Uwe Klammert
- Lehrstuhl für Mund-, Kiefer- und plastische Gesichtschirurgie, Universitätsklinikum Würzburg, Pleicherwall 2, 97070, Würzburg, Germany.
| | - Thomas D Mueller
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von- Sachs-Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs- Platz 2, D-97082, Würzburg, Germany.
| | - Tina V Hellmann
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von- Sachs-Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs- Platz 2, D-97082, Würzburg, Germany.
| | - Kristian K Wuerzler
- Lehrstuhl für Mund-, Kiefer- und plastische Gesichtschirurgie, Universitätsklinikum Würzburg, Pleicherwall 2, 97070, Würzburg, Germany.
| | - Alexander Kotzsch
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von- Sachs-Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs- Platz 2, D-97082, Würzburg, Germany.
| | - Anna Schliermann
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, D-97070, Würzburg, Germany.
| | - Werner Schmitz
- Lehrstuhl für Biochemie und Molekularbiologie, Theodor-Boveri-Institut für Biowissenschaften, Universität Würzburg, Am Hubland, 97074, Würzburg, Germany.
| | - Alexander C Kuebler
- Lehrstuhl für Mund-, Kiefer- und plastische Gesichtschirurgie, Universitätsklinikum Würzburg, Pleicherwall 2, 97070, Würzburg, Germany.
| | - Walter Sebald
- Lehrstuhl für Physiologische Chemie II, Theodor-Boveri-Institut für Biowissenschaften, Universität Würzburg, Am Hubland, 97074, Würzburg, Germany.
| | - Joachim Nickel
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, D-97070, Würzburg, Germany. .,Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Translationszentrum »Regenerative Therapien für Krebs- und Muskuloskelettale Erkrankungen« - Institutsteil Würzburg, Würzburg, Germany.
| |
Collapse
|
34
|
Glypican Is a Modulator of Netrin-Mediated Axon Guidance. PLoS Biol 2015; 13:e1002183. [PMID: 26148345 PMCID: PMC4493048 DOI: 10.1371/journal.pbio.1002183] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/15/2015] [Indexed: 11/19/2022] Open
Abstract
Netrin is a key axon guidance cue that orients axon growth during neural circuit formation. However, the mechanisms regulating netrin and its receptors in the extracellular milieu are largely unknown. Here we demonstrate that in Caenorhabditis elegans, LON-2/glypican, a heparan sulfate proteoglycan, modulates UNC-6/netrin signaling and may do this through interactions with the UNC-40/DCC receptor. We show that developing axons misorient in the absence of LON-2/glypican when the SLT-1/slit guidance pathway is compromised and that LON-2/glypican functions in both the attractive and repulsive UNC-6/netrin pathways. We find that the core LON-2/glypican protein, lacking its heparan sulfate chains, and secreted forms of LON-2/glypican are functional in axon guidance. We also find that LON-2/glypican functions from the epidermal substrate cells to guide axons, and we provide evidence that LON-2/glypican associates with UNC-40/DCC receptor-expressing cells. We propose that LON-2/glypican acts as a modulator of UNC-40/DCC-mediated guidance to fine-tune axonal responses to UNC-6/netrin signals during migration.
Collapse
|
35
|
Wang X, Hu R, Ge A, Hu L, Wang S, Feng X, Du W, Liu BF. Highly efficient microfluidic sorting device for synchronizing developmental stages of C. elegans based on deflecting electrotaxis. LAB ON A CHIP 2015; 15:2513-21. [PMID: 25963054 DOI: 10.1039/c5lc00354g] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
C. elegans as a powerful model organism has been widely used in fundamental biological studies. Many of these studies frequently need a large number of different stage-synchronized worms due to the stage-specific features of C. elegans among 4 distinct larval stages and the adult stage. In this work, we present an interesting and cost-effective microfluidic approach to realize simultaneous sorting of C. elegans of different developmental stages by deflecting electrotaxis. The microfluidic device was fabricated using PDMS consisting of symmetric sorting channels with specific angles, which was further hybridized to an agarose plate. While applying an electric field, different stages of C. elegans would crawl to the negative pore with different angles due to their deflecting electrotaxis. Thus, the worms were separated and synchronized by stages. lon-2 mutant was further used to study this electrotactic response and the results indicated that the body size plays a key role in determining the deflecting angle in matured adult worms. In addition to discriminating wild-type hermaphrodites, it could also be employed to sort mutants with abnormal development sizes and males. Therefore, our device provided a versatile and highly efficient platform for sorting C. elegans to meet the requirement of large numbers of different stage-synchronized worms. It can also be further used to investigate the neuronal basis of deflecting electrotaxis in worms.
Collapse
Affiliation(s)
- Xixian Wang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Promotion of bone morphogenetic protein signaling by tetraspanins and glycosphingolipids. PLoS Genet 2015; 11:e1005221. [PMID: 25978409 PMCID: PMC4433240 DOI: 10.1371/journal.pgen.1005221] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/14/2015] [Indexed: 02/08/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) belong to the transforming growth factor β (TGFβ) superfamily of secreted molecules. BMPs play essential roles in multiple developmental and homeostatic processes in metazoans. Malfunction of the BMP pathway can cause a variety of diseases in humans, including cancer, skeletal disorders and cardiovascular diseases. Identification of factors that ensure proper spatiotemporal control of BMP signaling is critical for understanding how this pathway is regulated. We have used a unique and sensitive genetic screen to identify the plasma membrane-localized tetraspanin TSP-21 as a key new factor in the C. elegans BMP-like “Sma/Mab” signaling pathway that controls body size and postembryonic M lineage development. We showed that TSP-21 acts in the signal-receiving cells and genetically functions at the ligand-receptor level. We further showed that TSP-21 can associate with itself and with two additional tetraspanins, TSP-12 and TSP-14, which also promote Sma/Mab signaling. TSP-12 and TSP-14 can also associate with SMA-6, the type I receptor of the Sma/Mab pathway. Finally, we found that glycosphingolipids, major components of the tetraspanin-enriched microdomains, are required for Sma/Mab signaling. Our findings suggest that the tetraspanin-enriched membrane microdomains are important for proper BMP signaling. As tetraspanins have emerged as diagnostic and prognostic markers for tumor progression, and TSP-21, TSP-12 and TSP-14 are all conserved in humans, we speculate that abnormal BMP signaling due to altered expression or function of certain tetraspanins may be a contributing factor to cancer development. The bone morphogenetic protein (BMP) signaling pathway is required for multiple developmental processes during metazoan development. Various diseases, including cancer, can result from mis-regulation of the BMP pathway. Thus, it is critical to identify factors that ensure proper regulation of BMP signaling. Using the nematode C. elegans, we have devised a highly specific and sensitive genetic screen to identify new modulators in the BMP pathway. Through this screen, we identified three conserved tetraspanin molecules as novel factors that function to promote BMP signaling in a living organism. We further showed that these three tetraspanins likely form a complex and function together with glycosphingolipids to promote BMP signaling. Recent studies have implicated several tetraspanins in cancer initiation, progression and metastasis in mammals. Our findings suggest that the involvement of tetraspanins in cancer may partially be due to their function in modulating the activity of BMP signaling.
Collapse
|
37
|
Wang X, Liu J, Zhu Z, Ou G. The heparan sulfate-modifying enzyme glucuronyl C5-epimerase HSE-5 controls Caenorhabditis elegans Q neuroblast polarization during migration. Dev Biol 2015; 399:306-14. [PMID: 25614236 DOI: 10.1016/j.ydbio.2015.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 01/05/2015] [Accepted: 01/10/2015] [Indexed: 10/24/2022]
Abstract
Directional cell migration is fundamental for neural development, and extracellular factors are pivotal for this process. Heparan sulfate proteoglycans (HSPGs) that carry long chains of differentially modified sugar residues contribute to extracellular matrix; however, the functions of HSPG in guiding cell migration remain elusive. Here, we used the Caenorhabditis elegans mutant pool from the Million Mutation Project and isolated a mutant allele of the heparan sulfate-modifying enzyme glucuronyl C5-epimerase HSE-5. Loss-of-function of this enzyme resulted in defective Q neuroblast migration. We showed that hse-5 controlled Q cell migration in a cell non-autonomous manner. By performing live cell imaging in hse-5 mutant animals, we found that hse-5 controlled initial polarization during Q neuroblast migration. Furthermore, our genetic epistasis analysis demonstrated that lon-2 might act downstream of hse-5. Finally, rescue of the hse-5 mutant phenotype by expression of human and mouse hse-5 homologs suggested a conserved function for this gene in neural development. Taken together, our results indicated that proper HSPG modification in the extracellular matrix by HSE-5 is essential for neuroblast polarity during migration.
Collapse
Affiliation(s)
- Xiangming Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China.
| | - Jianhong Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Zhiwen Zhu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
38
|
Khare SM, Awasthi A, Venkataraman V, Koushika SP. Colored polydimethylsiloxane micropillar arrays for high throughput measurements of forces applied by genetic model organisms. BIOMICROFLUIDICS 2015; 9:014111. [PMID: 25713693 PMCID: PMC4312341 DOI: 10.1063/1.4906905] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
Measuring forces applied by multi-cellular organisms is valuable in investigating biomechanics of their locomotion. Several technologies have been developed to measure such forces, for example, strain gauges, micro-machined sensors, and calibrated cantilevers. We introduce an innovative combination of techniques as a high throughput screening tool to assess forces applied by multiple genetic model organisms. First, we fabricated colored Polydimethylsiloxane (PDMS) micropillars where the color enhances contrast making it easier to detect and track pillar displacement driven by the organism. Second, we developed a semi-automated graphical user interface to analyze the images for pillar displacement, thus reducing the analysis time for each animal to minutes. The addition of color reduced the Young's modulus of PDMS. Therefore, the dye-PDMS composite was characterized using Yeoh's hyperelastic model and the pillars were calibrated using a silicon based force sensor. We used our device to measure forces exerted by wild type and mutant Caenorhabditis elegans moving on an agarose surface. Wild type C. elegans exert an average force of ∼1 μN on an individual pillar and a total average force of ∼7.68 μN. We show that the middle of C. elegans exerts more force than its extremities. We find that C. elegans mutants with defective body wall muscles apply significantly lower force on individual pillars, while mutants defective in sensing externally applied mechanical forces still apply the same average force per pillar compared to wild type animals. Average forces applied per pillar are independent of the length, diameter, or cuticle stiffness of the animal. We also used the device to measure, for the first time, forces applied by Drosophila melanogaster larvae. Peristaltic waves occurred at 0.4 Hz applying an average force of ∼1.58 μN on a single pillar. Our colored microfluidic device along with its displacement tracking software allows us to measure forces applied by multiple model organisms that crawl or slither to travel through their environment.
Collapse
Affiliation(s)
- Siddharth M Khare
- Department of Physics, Indian Institute of Science , Bangalore 560012, India
| | | | - V Venkataraman
- Department of Physics, Indian Institute of Science , Bangalore 560012, India
| | | |
Collapse
|
39
|
Dineen A, Gaudet J. TGF-β signaling can act from multiple tissues to regulate C. elegans body size. BMC DEVELOPMENTAL BIOLOGY 2014; 14:43. [PMID: 25480452 PMCID: PMC4278669 DOI: 10.1186/s12861-014-0043-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 11/25/2014] [Indexed: 11/23/2022]
Abstract
Background Regulation of organ and body size is a fundamental biological phenomenon, requiring tight coordination between multiple tissues to ensure accurate proportional growth. In C. elegans, a TGF-β pathway is the major regulator of body size and also plays a role in the development of the male tail, and is thus referred to as the TGF-β/Sma/Mab (for small and male abnormal) pathway. Mutations in components of this pathway result in decreased growth of animals during larval stages, with Sma mutant adults of the core pathway as small as ~60-70% the length of normal animals. The currently accepted model suggests that TGF-β/Sma/Mab pathway signaling in the C. elegans hypodermis is both necessary and sufficient to control body length. However, components of this signaling pathway are expressed in other organs, such as the intestine and pharynx, raising the question of what the function of the pathway is in these organs. Results Here we show that TGF-β/Sma/Mab signaling is required for the normal growth of the pharynx. We further extend the current model and show that the TGF-β/Sma/Mab pathway can function in multiple tissues to regulate body and organ length. Specifically, we find that pharyngeal expression of the SMAD protein SMA-3 partially rescues both pharynx length and body length of sma-3 mutants. Conclusions Overall, our results support a model in which the TGF-β/Sma/Mab signaling pathway can act in multiple tissues, activating one or more downstream secreted signals that act non cell-autonomously to regulate overall body length in C. elegans. Electronic supplementary material The online version of this article (doi:10.1186/s12861-014-0043-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aidan Dineen
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4 N1, Alberta, Canada.
| | - Jeb Gaudet
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4 N1, Alberta, Canada.
| |
Collapse
|
40
|
Shen Z, Zhang X, Chai Y, Zhu Z, Yi P, Feng G, Li W, Ou G. Conditional knockouts generated by engineered CRISPR-Cas9 endonuclease reveal the roles of coronin in C. elegans neural development. Dev Cell 2014; 30:625-36. [PMID: 25155554 DOI: 10.1016/j.devcel.2014.07.017] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 07/07/2014] [Accepted: 07/21/2014] [Indexed: 10/24/2022]
Abstract
Conditional gene knockout animals are valuable tools for studying the mechanisms underlying cell and developmental biology. We developed a conditional knockout strategy by spatiotemporally manipulating the expression of an RNA-guided DNA endonuclease, CRISPR-Cas9, in Caenorhabditis elegans somatic cell lineages. We showed that this somatic CRISPR-Cas9 technology provides a quick and efficient approach to generate conditional knockouts in various cell types at different developmental stages. Furthermore, we demonstrated that this method outperforms our recently developed somatic TALEN technique and enables the one-step generation of multiple conditional knockouts. By combining these techniques with live-cell imaging, we showed that an essential embryonic gene, Coronin, which is associated with human neurobehavioral dysfunction, regulates actin organization and cell morphology during C. elegans postembryonic neuroblast migration and neuritogenesis. We propose that the somatic CRISPR-Cas9 platform is uniquely suited for conditional gene editing-based biomedical research.
Collapse
Affiliation(s)
- Zhongfu Shen
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xianliang Zhang
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yongping Chai
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhiwen Zhu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Peishan Yi
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Guoxin Feng
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Li
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
41
|
Kinnunen TK. Combinatorial roles of heparan sulfate proteoglycans and heparan sulfates in Caenorhabditis elegans neural development. PLoS One 2014; 9:e102919. [PMID: 25054285 PMCID: PMC4108370 DOI: 10.1371/journal.pone.0102919] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 06/25/2014] [Indexed: 02/06/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) play critical roles in the development and adult physiology of all metazoan organisms. Most of the known molecular interactions of HSPGs are attributed to the structurally highly complex heparan sulfate (HS) glycans. However, whether a specific HSPG (such as syndecan) contains HS modifications that differ from another HSPG (such as glypican) has remained largely unresolved. Here, a neural model in C. elegans is used to demonstrate for the first time the relationship between specific HSPGs and HS modifications in a defined biological process in vivo. HSPGs are critical for the migration of hermaphrodite specific neurons (HSNs) as genetic elimination of multiple HSPGs leads to 80% defect of HSN migration. The effects of genetic elimination of HSPGs are additive, suggesting that multiple HSPGs, present in the migrating neuron and in the matrix, act in parallel to support neuron migration. Genetic analyses suggest that syndecan/sdn-1 and HS 6-O-sulfotransferase, hst-6, function in a linear signaling pathway and glypican/lon-2 and HS 2-O-sulfotransferase, hst-2, function together in a pathway that is parallel to sdn-1 and hst-6. These results suggest core protein specific HS modifications that are critical for HSN migration. In C. elegans, the core protein specificity of distinct HS modifications may be in part regulated at the level of tissue specific expression of genes encoding for HSPGs and HS modifying enzymes. Genetic analysis reveals that there is a delicate balance of HS modifications and eliminating one HS modifying enzyme in a compromised genetic background leads to significant changes in the overall phenotype. These findings are of importance with the view of HS as a critical regulator of cell signaling in normal development and disease.
Collapse
Affiliation(s)
- Tarja K. Kinnunen
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom, and Department of Biology, University of Huddersfield, Huddersfield, United Kingdom
- * E-mail:
| |
Collapse
|
42
|
Petzold BC, Park SJ, Mazzochette EA, Goodman MB, Pruitt BL. MEMS-based force-clamp analysis of the role of body stiffness in C. elegans touch sensation. Integr Biol (Camb) 2014; 5:853-64. [PMID: 23598612 DOI: 10.1039/c3ib20293c] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Touch is enabled by mechanoreceptor neurons in the skin and plays an essential role in our everyday lives, but is among the least understood of our five basic senses. Force applied to the skin deforms these neurons and activates ion channels within them. Despite the importance of the mechanics of the skin in determining mechanoreceptor neuron deformation and ultimately touch sensation, the role of mechanics in touch sensitivity is poorly understood. Here, we use the model organism Caenorhabditis elegans to directly test the hypothesis that body mechanics modulate touch sensitivity. We demonstrate a microelectromechanical system (MEMS)-based force clamp that can apply calibrated forces to freely crawling C. elegans worms and measure touch-evoked avoidance responses. This approach reveals that wild-type animals sense forces <1 μN and indentation depths <1 μm. We use both genetic manipulation of the skin and optogenetic modulation of body wall muscles to alter body mechanics. We find that small changes in body stiffness dramatically affect force sensitivity, while having only modest effects on indentation sensitivity. We investigate the theoretical body deformation predicted under applied force and conclude that local mechanical loads induce inward bending deformation of the skin to drive touch sensation in C. elegans.
Collapse
Affiliation(s)
- Bryan C Petzold
- Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California, USA
| | | | | | | | | |
Collapse
|
43
|
Neumann B, Hilliard MA. Loss of MEC-17 leads to microtubule instability and axonal degeneration. Cell Rep 2013; 6:93-103. [PMID: 24373971 DOI: 10.1016/j.celrep.2013.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 10/31/2013] [Accepted: 12/03/2013] [Indexed: 10/25/2022] Open
Abstract
Axonal degeneration arises as a consequence of neuronal injury and is a common hallmark of a number of neurodegenerative diseases. However, the genetic causes and the cellular mechanisms that trigger this process are still largely unknown. Based on forward genetic screening in C. elegans, we have identified the α-tubulin acetyltransferase gene mec-17 as causing spontaneous, adult-onset, and progressive axonal degeneration. Loss of MEC-17 leads to microtubule instability, a reduction in mitochondrial number, and disrupted axonal transport, with altered distribution of both mitochondria and synaptic components. Furthermore, mec-17-mediated axonal degeneration occurs independently from its acetyltransferase domain; is enhanced by mutation of coel-1, a tubulin-associated molecule; and correlates with the animal's body length. This study therefore identifies a critical role for the conserved microtubule-associated protein MEC-17 in preserving axon integrity and preventing axonal degeneration.
Collapse
Affiliation(s)
- Brent Neumann
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo A Hilliard
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
44
|
Tuck S. The control of cell growth and body size in Caenorhabditis elegans. Exp Cell Res 2013; 321:71-6. [PMID: 24262077 DOI: 10.1016/j.yexcr.2013.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 01/02/2023]
Abstract
One of the most important ways in which animal species vary is in their size. Individuals of the largest animal ever thought to have lived, the blue whale (Balaenoptera musculus), can reach a weight of 190 t and a length of over 30 m. At the other extreme, among the smallest multicellular animals are males of the parasitic wasp, Dicopomorpha echmepterygis, which even as adults are just 140 μm in length. In terms of volume, these species differ by more than 14 orders of magnitude. Since size has such profound effects on an organism's ecology, anatomy and physiology, an important task for evolutionary biology and ecology is to account for why organisms grow to their characteristic sizes. Equally, a full description of an organism's development must include an explanation of how its growth and body size are regulated. Here I review research on how these processes are controlled in the nematode, Caenorhabditis elegans. Analyses of small and long mutants have revealed that in the worm, DBL-1, a ligand in the TGFβ superfamily family, promotes growth in a dose-dependent manner. DBL-1 signaling affects body size by stimulating the growth of syncytial hypodermal cells rather than controlling cell division. Signals from chemosensory neurons and from the gonad also modulate body size, in part, independently of DBL-1-mediated signaling. Organismal size and morphology is heavily influenced by the cuticle, which acts as the exoskeleton. Finally, I summarize research on several genes that appear to regulate body size by cell autonomously regulating cell growth throughout the worm.
Collapse
Affiliation(s)
- Simon Tuck
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
45
|
Pedersen ME, Snieckute G, Kagias K, Nehammer C, Multhaupt HAB, Couchman JR, Pocock R. An epidermal microRNA regulates neuronal migration through control of the cellular glycosylation state. Science 2013; 341:1404-8. [PMID: 24052309 DOI: 10.1126/science.1242528] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
An appropriate balance in glycosylation of proteoglycans is crucial for their ability to regulate animal development. Here, we report that the Caenorhabditis elegans microRNA mir-79, an ortholog of mammalian miR-9, controls sugar-chain homeostasis by targeting two proteins in the proteoglycan biosynthetic pathway: a chondroitin synthase (SQV-5; squashed vulva-5) and a uridine 5'-diphosphate-sugar transporter (SQV-7). Loss of mir-79 causes neurodevelopmental defects through SQV-5 and SQV-7 dysregulation in the epidermis. This results in a partial shutdown of heparan sulfate biosynthesis that impinges on a LON-2/glypican pathway and disrupts neuronal migration. Our results identify a regulatory axis controlled by a conserved microRNA that maintains proteoglycan homeostasis in cells.
Collapse
Affiliation(s)
- Mikael Egebjerg Pedersen
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
46
|
Cheng Z, Yi P, Wang X, Chai Y, Feng G, Yang Y, Liang X, Zhu Z, Li W, Ou G. Conditional targeted genome editing using somatically expressed TALENs in C. elegans. Nat Biotechnol 2013; 31:934-7. [PMID: 23955274 DOI: 10.1038/nbt.2674] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/22/2013] [Indexed: 11/08/2022]
Abstract
We have developed a method for the generation of conditional knockouts in Caenorhabditis elegans by expressing transcription activator-like effector nucleases (TALENs) in somatic cells. Using germline transformation with plasmids encoding TALENs under the control of an inducible or tissue-specific promoter, we observed effective gene modifications and resulting phenotypes in specific developmental stages and tissues. We further used this method to bypass the embryonic requirement of cor-1, which encodes the homolog of human severe combined immunodeficiency (SCID) protein coronin, and we determined its essential role in cell migration in larval Q-cell lineages. Our results show that TALENs expressed in the somatic cells of model organisms provide a versatile tool for functional genomics.
Collapse
Affiliation(s)
- Ze Cheng
- 1] National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China. [2] School of Life Sciences, Peking University, Beijing, China. [3]
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Taneja-Bageshwar S, Gumienny TL. Regulation of TGFβ superfamily signaling by two separable domains of glypican LON-2 in C. elegans. WORM 2013; 2:e23843. [PMID: 24778932 PMCID: PMC3875644 DOI: 10.4161/worm.23843] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/31/2013] [Indexed: 12/30/2022]
Abstract
Regulated intercellular signaling is critical for the normal development and maintenance of multicellular organisms. Glypicans have been shown to regulate signaling by TGFβs, hedgehogs and Wnts, in several cellular contexts. Glypicans comprise a conserved family of heparan sulfated, glycosylphosphatidylinositol (GPI)-linked extracellular proteins. The structural complexity of glypicans may underlie their functional complexity. In a recent study31, we built on previous findings that one of the two C. elegans glypicans, LON-2, specifically inhibits signaling by the TGFβ superfamily member DBL-1. We tested the functional requirements of LON-2 protein core components and post-translational modifications for LON-2 activity. We provide the first evidence that two parts of a glypican can independently regulate TGFβ superfamily signaling in vivo: the N-terminal furin protease product and a C-terminal region containing heparan sulfate attachment sites. Furthermore, we show a protein-protein interaction motif is crucial for LON-2 activity in the N-terminal protein core, suggesting that LON-2 acts by serving as a scaffold for DBL-1 and an RGD-binding protein. In addition, we demonstrate specificity of glypican function by showing C. elegans GPN-1 does not functionally substitute for LON-2. This work reveals a molecular foundation for understanding the complexity and specificity of glypican function.
Collapse
Affiliation(s)
- Suparna Taneja-Bageshwar
- Department of Molecular and Cellular Medicine; College of Medicine; Texas A&M Health Science Center; College Station, TX USA
| | - Tina L Gumienny
- Department of Molecular and Cellular Medicine; College of Medicine; Texas A&M Health Science Center; College Station, TX USA
| |
Collapse
|
48
|
Gysi S, Rhiner C, Flibotte S, Moerman DG, Hengartner MO. A network of HSPG core proteins and HS modifying enzymes regulates netrin-dependent guidance of D-type motor neurons in Caenorhabditis elegans. PLoS One 2013; 8:e74908. [PMID: 24066155 PMCID: PMC3774775 DOI: 10.1371/journal.pone.0074908] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 08/07/2013] [Indexed: 11/18/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are proteins with long covalently attached sugar side chains of the heparan sulfate (HS) type. Depending on the cellular context HS chains carry multiple structural modifications such as sulfate residues or epimerized sugars allowing them to bind to a wide range of molecules. HSPGs have been found to play extremely diverse roles in animal development and were shown to interact with certain axon guidance molecules. In this study we describe the role of the Caenorhabditis elegans HSPG core proteins Syndecan (SDN-1) and Glypican (LON-2) and the HS modifying enzymes in the dorsal guidance of D-type motor axons, a process controlled mainly by the conserved axon guidance molecule UNC-6/Netrin. Our genetic analysis established the specific HS code relevant for this axon guidance event. Using two sensitized genetic backgrounds, we isolated novel components influencing D-type motor axon guidance with a link to HSPGs, as well as new alleles of several previously characterized axon guidance genes. Interestingly, the dorsal axon guidance defects induced by mutations in zfp-1 or lin-35 depended on the transgene oxIs12 used to visualize the D-type motor neurons. oxIs12 is a large multi-copy transgene that enlarges the X chromosome by approximately 20%. In a search for genes with a comparable phenotype we found that a mutation in the known dosage compensation gene dpy-21 showed similar axon guidance defects as zfp-1 or lin-35 mutants. Thus, derepression of genes on X, where many genes relevant for HS dependent axon guidance are located, might also influence axon guidance of D-type motor neurons.
Collapse
Affiliation(s)
- Stephan Gysi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Christa Rhiner
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, Canada
| | - Donald G. Moerman
- Department of Zoology, University of British Columbia, Vancouver, Canada
| | - Michael O. Hengartner
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Tian C, Shi H, Xiong S, Hu F, Xiong WC, Liu J. The neogenin/DCC homolog UNC-40 promotes BMP signaling via the RGM protein DRAG-1 in C. elegans. Development 2013; 140:4070-80. [PMID: 24004951 DOI: 10.1242/dev.099838] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The deleted in colorectal cancer (DCC) homolog neogenin functions in both netrin- and repulsive guidance molecule (RGM)-mediated axon guidance and in bone morphogenetic protein (BMP) signaling. How neogenin functions in mediating BMP signaling is not well understood. We show that the sole C. elegans DCC/neogenin homolog UNC-40 positively modulates a BMP-like pathway by functioning in the signal-receiving cells at the ligand/receptor level. This function of UNC-40 is independent of its role in netrin-mediated axon guidance, but requires its association with the RGM protein DRAG-1. We have identified the key residues in the extracellular domain of UNC-40 that are crucial for UNC-40-DRAG-1 interaction and UNC-40 function. Surprisingly, the extracellular domain of UNC-40 is sufficient to promote BMP signaling, in clear contrast to the requirement of its intracellular domain in mediating axon guidance. Mouse neogenin lacking the intracellular domain is also capable of mediating BMP signaling. These findings reveal an unexpected mode of action for neogenin regulation of BMP signaling.
Collapse
Affiliation(s)
- Chenxi Tian
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | |
Collapse
|
50
|
Tian C, Liu J. Repulsive guidance molecules (RGMs) and neogenin in bone morphogenetic protein (BMP) signaling. Mol Reprod Dev 2013; 80:700-17. [PMID: 23740870 DOI: 10.1002/mrd.22199] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/28/2013] [Indexed: 02/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-beta (TGFβ) superfamily. BMPs mediate a highly conserved signal transduction cascade through the type-I and type-II serine/threonine kinase receptors and intracellular Smad proteins, which regulate multiple developmental and homeostatic processes. Mutations in this pathway can cause various diseases in humans, such as skeletal disorders, cardiovascular diseases, and various cancers. Multiple levels of regulation, including extracellular regulation, help to ensure proper spatiotemporal control of BMP signaling in the right cellular context. The family of repulsive guidance molecules (RGMs) and the type-I transmembrane protein neogenin, a paralog of DCC (Deleted in Colorectal Cancer), have been implicated in modulating the BMP pathway. In this review, we discuss the properties and functions of RGM proteins and neogenin, focusing on their roles in the modulation of BMP signal transduction.
Collapse
Affiliation(s)
- Chenxi Tian
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York
| | | |
Collapse
|