1
|
Vora M, Dietz J, Wing Z, 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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597576. [PMID: 38895257 PMCID: PMC11185707 DOI: 10.1101/2024.06.05.597576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
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, Dept. of Genetics, Rutgers University, NJ, USA
- ModOmics Ltd, Southampton, UK
| | - Jonathan Dietz
- Waksman Institute, Dept. of Genetics, Rutgers University, NJ, USA
| | - Zachary Wing
- Department of Biology, Queens College, CUNY, NY, USA
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, NY, USA
| | | | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY, NY, USA
- PhD Program in Biology, the Graduate Center, CUNY, NY, USA
| |
Collapse
|
2
|
Ciccarelli EJ, Bendelstein M, Yamamoto KK, Reich H, Savage-Dunn C. BMP signaling to pharyngeal muscle in the C. elegans response to a bacterial pathogen regulates anti-microbial peptide expression and pharyngeal pumping. Mol Biol Cell 2024; 35:ar52. [PMID: 38381557 PMCID: PMC11064665 DOI: 10.1091/mbc.e23-05-0185] [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: 05/24/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024] Open
Abstract
Host response to pathogens recruits multiple tissues in part through conserved cell signaling pathways. In Caenorhabditis elegans, the bone morphogenetic protein (BMP) like DBL-1 signaling pathway has a role in the response to infection in addition to other roles in development and postdevelopmental functions. In the regulation of body size, the DBL-1 pathway acts through cell autonomous signal activation in the epidermis (hypodermis). We have now elucidated the tissues that respond to DBL-1 signaling upon exposure to two bacterial pathogens. The receptors and Smad signal transducers for DBL-1 are expressed in pharyngeal muscle, intestine, and epidermis. We demonstrate that expression of receptor-regulated Smad (R-Smad) gene sma-3 in the pharynx is sufficient to improve the impaired survival phenotype of sma-3 mutants and that expression of sma-3 in the intestine has no effect when exposing worms to bacterial infection of the intestine. We also show that two antimicrobial peptide genes - abf-2 and cnc-2 - are regulated by DBL-1 signaling through R-Smad SMA-3 activity in the pharynx. Finally, we show that pharyngeal pumping activity is reduced in sma-3 mutants and that other pharynx-defective mutants also have reduced survival on a bacterial pathogen. Our results identify the pharynx as a tissue that responds to BMP signaling to coordinate a systemic response to bacterial pathogens.
Collapse
Affiliation(s)
- Emma Jo Ciccarelli
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| | | | - Katerina K. Yamamoto
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| | - Hannah Reich
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| |
Collapse
|
3
|
Ciccarelli EJ, Bendelstein M, Yamamoto KK, Reich H, Savage-Dunn C. BMP signaling to pharyngeal muscle in the C. elegans response to a bacterial pathogen regulates anti-microbial peptide expression and pharyngeal pumping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.06.531324. [PMID: 36945421 PMCID: PMC10028841 DOI: 10.1101/2023.03.06.531324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Host response to pathogens recruits multiple tissues in part through conserved cell signaling pathways. In C. elegans, the bone morphogenetic protein (BMP) like DBL-1 signaling pathway has a role in the response to infection in addition to other roles in development and post-developmental functions. In the regulation of body size, the DBL-1 pathway acts through cell autonomous signal activation in the epidermis (hypodermis). We have now elucidated the tissues that respond to DBL-1 signaling upon exposure to two bacterial pathogens. The receptors and Smad signal transducers for DBL-1 are expressed in pharyngeal muscle, intestine, and epidermis. We demonstrate that expression of receptor-regulated Smad (R-Smad) gene sma-3 in the pharynx is sufficient to improve the impaired survival phenotype of sma-3 mutants and that expression of sma-3 in the intestine has no effect when exposing worms to bacterial infection of the intestine. We also show that two antimicrobial peptide genes - abf-2 and cnc-2 - are regulated by DBL-1 signaling through R-Smad SMA-3 activity in the pharynx. Finally, we show that pharyngeal pumping activity is reduced in sma-3 mutants and that other pharynx-defective mutants also have reduced survival on a bacterial pathogen. Our results identify the pharynx as a tissue that responds to BMP signaling to coordinate a systemic response to bacterial pathogens.
Collapse
Affiliation(s)
- Emma Jo Ciccarelli
- Department of Biology, Queens College, CUNY, Flushing NY
- PhD Program in Biology, The Graduate Center, CUNY, New York NY
| | | | - Katerina K. Yamamoto
- Department of Biology, Queens College, CUNY, Flushing NY
- PhD Program in Biology, The Graduate Center, CUNY, New York NY
| | - Hannah Reich
- Department of Biology, Queens College, CUNY, Flushing NY
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY, Flushing NY
- PhD Program in Biology, The Graduate Center, CUNY, New York NY
| |
Collapse
|
4
|
Taylor SKB, Hartman JH, Gupta BP. Neurotrophic factor MANF regulates autophagy and lysosome function to promote proteostasis in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.31.551399. [PMID: 38260421 PMCID: PMC10802257 DOI: 10.1101/2023.07.31.551399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The conserved mesencephalic astrocyte-derived neurotrophic factor (MANF) protects dopaminergic neurons but also functions in several other tissues. Previously, we showed that Caenorhabditis elegans manf-1 null mutants have increased ER stress, dopaminergic neurodegeneration, protein aggregation, slower growth, and a reduced lifespan. The multiple requirements of MANF in different systems suggest its essential role in regulating cellular processes. However, how intracellular and extracellular MANF regulates broader cellular function remains unknown. Here, we report a novel mechanism of action for manf-1 that involves the autophagy transcription factor HLH-30/TFEB-mediated signaling to regulate lysosomal function and aging. We generated multiple transgenic strains overexpressing MANF-1 and found that animals had extended lifespan, reduced protein aggregation, and improved neuronal health. Using a fluorescently tagged MANF-1, we observed different tissue localization of MANF-1 depending on the ER retention signal. Further subcellular analysis showed that MANF-1 localizes within cells to the lysosomes. These findings were consistent with our transcriptomic studies and, together with analysis of autophagy regulators, demonstrate that MANF-1 regulates protein homeostasis through increased autophagy and lysosomal activity. Collectively, our findings establish MANF as a critical regulator of the stress response, proteostasis, and aging.
Collapse
Affiliation(s)
- Shane K. B. Taylor
- Department of Biology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jessica H. Hartman
- Department of Biochemistry & Molecular Biology and Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bhagwati P. Gupta
- Department of Biology, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
5
|
Chan WT, Medriano CA, Bae S. Unveiling the impact of short-term polyethylene microplastics exposure on metabolomics and gut microbiota in earthworms (Eudrilus euganiae). JOURNAL OF HAZARDOUS MATERIALS 2023; 460:132305. [PMID: 37672993 DOI: 10.1016/j.jhazmat.2023.132305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 09/08/2023]
Abstract
Microplastics (MPs) pose a significant environmental concern, particularly for terrestrial fauna. In this study, earthworms were used as a model organism to investigate the ecotoxicological effects of short-term exposure to virgin MPs on changes in metabolome and gut microbiota. High-throughput untargeted metabolomics showed significant internal reactions in the earthworms' metabolic processes due to MPs exposure, even when no visible stress signs, such as changes in growth or mortality rates, were present. Earthworms exposed to different concentrations of polyethylene (PE) MP exhibited significant disruption in 39 and 199 molecular features related to energy and lipid metabolism, anti-inflammatory, cell signaling, and membrane integrity. The activities of enzymes and transport proteins in earthworms were dysregulated when exposed to PE. Changes in the gut microbiota's community structure and complexity were observed in response to PE MPs exposure. Despite the relative stability in alpha-diversity and relative abundance, shifts in beta-diversity and network analysis in the PE-exposed group were indicative of an adaptive response to MPs. Earthworms exhibited resilience or adaptation in response to MPs exposure, potentially maintaining their functionality. This study provides preliminary insights into the impact of MPs on soil invertebrates like earthworms and highlights the need for further exploration of long-term effects and underlying molecular mechanisms.
Collapse
Affiliation(s)
- Wan Ting Chan
- Department of Civil and Environmental Engineering, National University of Singapore, 117576, Singapore
| | - Carl Angelo Medriano
- Department of Civil and Environmental Engineering, National University of Singapore, 117576, Singapore
| | - Sungwoo Bae
- Department of Civil and Environmental Engineering, National University of Singapore, 117576, Singapore.
| |
Collapse
|
6
|
Madhu B, Lakdawala MF, Gumienny TL. The DBL-1/TGF-β signaling pathway tailors behavioral and molecular host responses to a variety of bacteria in Caenorhabditis elegans. eLife 2023; 12:e75831. [PMID: 37750680 PMCID: PMC10567113 DOI: 10.7554/elife.75831] [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: 12/07/2021] [Accepted: 09/25/2023] [Indexed: 09/27/2023] Open
Abstract
Generating specific, robust protective responses to different bacteria is vital for animal survival. Here, we address the role of transforming growth factor β (TGF-β) member DBL-1 in regulating signature host defense responses in Caenorhabditis elegans to human opportunistic Gram-negative and Gram-positive pathogens. Canonical DBL-1 signaling is required to suppress avoidance behavior in response to Gram-negative, but not Gram-positive bacteria. We propose that in the absence of DBL-1, animals perceive some bacteria as more harmful. Animals activate DBL-1 pathway activity in response to Gram-negative bacteria and strongly repress it in response to select Gram-positive bacteria, demonstrating bacteria-responsive regulation of DBL-1 signaling. DBL-1 signaling differentially regulates expression of target innate immunity genes depending on the bacterial exposure. These findings highlight a central role for TGF-β in tailoring a suite of bacteria-specific host defenses.
Collapse
Affiliation(s)
- Bhoomi Madhu
- Department of Biology, Texas Woman’s UniversityDentonUnited States
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Mohammed Farhan Lakdawala
- Department of Biology, Texas Woman’s UniversityDentonUnited States
- AbbVie (United States)WorcesterUnited States
| | - Tina L Gumienny
- Department of Biology, Texas Woman’s UniversityDentonUnited States
| |
Collapse
|
7
|
Yamamoto KK, Savage-Dunn C. TGF-β pathways in aging and immunity: lessons from Caenorhabditis elegans. Front Genet 2023; 14:1220068. [PMID: 37732316 PMCID: PMC10507863 DOI: 10.3389/fgene.2023.1220068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/23/2023] [Indexed: 09/22/2023] Open
Abstract
The Transforming Growth Factor-β (TGF-β) superfamily of signaling molecules plays critical roles in development, differentiation, homeostasis, and disease. Due to the conservation of these ligands and their signaling pathways, genetic studies in invertebrate systems including the nematode Caenorhabditis elegans have been instrumental in identifying signaling mechanisms. C. elegans is also a premier organism for research in longevity and healthy aging. Here we summarize current knowledge on the roles of TGF-β signaling in aging and immunity.
Collapse
Affiliation(s)
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, and PhD Program in Biology, The Graduate Center, City University of New York, New York City, NY, United States
| |
Collapse
|
8
|
Li Q, Xiao M, Li N, Cai W, Zhao C, Liu B, Zeng F. Application of
Caenorhabditis elegans
in the evaluation of food nutrition: A review. EFOOD 2023. [DOI: 10.1002/efd2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Affiliation(s)
- Quancen Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Meifang Xiao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Wenwen Cai
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| | - Bin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou China
| | - Feng Zeng
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
9
|
Lo WS, Roca M, Dardiry M, Mackie M, Eberhardt G, Witte H, Hong R, Sommer RJ, Lightfoot JW. Evolution and Diversity of TGF-β Pathways are Linked with Novel Developmental and Behavioral Traits. Mol Biol Evol 2022; 39:msac252. [PMID: 36469861 PMCID: PMC9733428 DOI: 10.1093/molbev/msac252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is essential for numerous biologic functions. It is a highly conserved pathway found in all metazoans including the nematode Caenorhabditis elegans, which has also been pivotal in identifying many components. Utilizing a comparative evolutionary approach, we explored TGF-β signaling in nine nematode species and revealed striking variability in TGF-β gene frequency across the lineage. Of the species analyzed, gene duplications in the DAF-7 pathway appear common with the greatest disparity observed in Pristionchus pacificus. Specifically, multiple paralogues of daf-3, daf-4 and daf-7 were detected. To investigate this additional diversity, we induced mutations in 22 TGF-β components and generated corresponding double, triple, and quadruple mutants revealing both conservation and diversification in function. Although the DBL-1 pathway regulating body morphology appears highly conserved, the DAF-7 pathway exhibits functional divergence, notably in some aspects of dauer formation. Furthermore, the formation of the phenotypically plastic mouth in P. pacificus is partially influenced through TGF-β with the strongest effect in Ppa-tag-68. This appears important for numerous processes in P. pacificus but has no known function in C. elegans. Finally, we observe behavioral differences in TGF-β mutants including in chemosensation and the establishment of the P. pacificus kin-recognition signal. Thus, TGF-β signaling in nematodes represents a stochastic genetic network capable of generating novel functions through the duplication and deletion of associated genes.
Collapse
Affiliation(s)
- Wen-Sui Lo
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology Tübingen, Max-Planck Ring 9, 72076 Tübingen, Germany
| | - Marianne Roca
- Max Planck Research Group Genetics of Behavior, Max Planck Institute for Neurobiology of Behavior—Caesar, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Mohannad Dardiry
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology Tübingen, Max-Planck Ring 9, 72076 Tübingen, Germany
| | - Marisa Mackie
- Department of Biology, California State University, Northridge, CA
| | - Gabi Eberhardt
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology Tübingen, Max-Planck Ring 9, 72076 Tübingen, Germany
| | - Hanh Witte
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology Tübingen, Max-Planck Ring 9, 72076 Tübingen, Germany
| | - Ray Hong
- Department of Biology, California State University, Northridge, CA
| | - Ralf J Sommer
- Department for Integrative Evolutionary Biology, Max-Planck Institute for Biology Tübingen, Max-Planck Ring 9, 72076 Tübingen, Germany
| | - James W Lightfoot
- Max Planck Research Group Genetics of Behavior, Max Planck Institute for Neurobiology of Behavior—Caesar, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| |
Collapse
|
10
|
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
|
11
|
Chomyshen SC, Tabarraei H, Wu CW. Translational suppression via IFG-1/eIF4G inhibits stress-induced RNA alternative splicing in Caenorhabditis elegans. Genetics 2022; 221:iyac075. [PMID: 35536193 PMCID: PMC9252287 DOI: 10.1093/genetics/iyac075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Splicing of precursor mRNA is an essential process for dividing cells, and splicing defects have been linked to aging and various chronic diseases. Environmental stress has recently been shown to modify alternative splicing, and molecular mechanisms that influence stress-induced alternative splicing remain unclear. Using an in vivo RNA splicing reporter, we performed a genome-wide RNAi screen in Caenorhabditis elegans and found that protein translation suppression via silencing of the conserved eukaryotic initiation factor 4G (IFG-1/eIF4G) inhibits cadmium-induced alternative splicing. Transcriptome analysis of an ifg-1-deficient mutant revealed an overall decrease in intronic and intergenic reads and prevented cadmium-induced alternative splicing compared to the wild type. We found that the ifg-1 mutant up-regulates >80 RNA splicing regulatory genes controlled by the TGF-β transcription factor SMA-2. The extended lifespan of the ifg-1 mutant is partially reduced upon sma-2 depletion and completely nullified when core spliceosome genes including snr-1, snr-2, and uaf-2 are knocked down. Depletion of snr-1 and snr-2 also diminished the enhanced cadmium resistance of the ifg-1 mutant. Together, these data describe a molecular mechanism through which translation suppression inhibits stress-induced alternative splicing and demonstrate an essential role for RNA splicing in promoting longevity and stress resistance in a translation-compromised mutant.
Collapse
Affiliation(s)
- Samantha C Chomyshen
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Hadi Tabarraei
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Cheng-Wei Wu
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
12
|
Baltaci O, Pedersen ME, Sherry T, Handley A, Snieckute G, Cao W, Haas M, Archer S, Pocock R. Atypical TGF-β signaling controls neuronal guidance in Caenorhabditis elegans. iScience 2022; 25:103791. [PMID: 35146399 PMCID: PMC8819019 DOI: 10.1016/j.isci.2022.103791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/09/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022] Open
Abstract
Coordinated expression of cell adhesion and signaling molecules is crucial for brain development. Here, we report that the Caenorhabditis elegans transforming growth factor β (TGF-β) type I receptor SMA-6 (small-6) acts independently of its cognate TGF-β type II receptor DAF-4 (dauer formation-defective-4) to control neuronal guidance. SMA-6 directs neuronal development from the hypodermis through interactions with three, orphan, TGF-β ligands. Intracellular signaling downstream of SMA-6 limits expression of NLR-1, an essential Neurexin-like cell adhesion receptor, to enable neuronal guidance. Together, our data identify an atypical TGF-β-mediated regulatory mechanism to ensure correct neuronal development.
Collapse
Affiliation(s)
- Oguzhan Baltaci
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Mikael Egebjerg Pedersen
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, Denmark
| | - Tessa Sherry
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Ava Handley
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Goda Snieckute
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, Denmark
| | - Wei Cao
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Matilda Haas
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Stuart Archer
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC 3800, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, Denmark
| |
Collapse
|
13
|
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
|
14
|
Chauve L, Hodge F, Murdoch S, Masoudzadeh F, Mann HJ, Lopez-Clavijo AF, Okkenhaug H, West G, Sousa BC, Segonds-Pichon A, Li C, Wingett SW, Kienberger H, Kleigrewe K, de Bono M, Wakelam MJO, Casanueva O. Neuronal HSF-1 coordinates the propagation of fat desaturation across tissues to enable adaptation to high temperatures in C. elegans. PLoS Biol 2021; 19:e3001431. [PMID: 34723964 PMCID: PMC8585009 DOI: 10.1371/journal.pbio.3001431] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 11/11/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
To survive elevated temperatures, ectotherms adjust the fluidity of membranes by fine-tuning lipid desaturation levels in a process previously described to be cell autonomous. We have discovered that, in Caenorhabditis elegans, neuronal heat shock factor 1 (HSF-1), the conserved master regulator of the heat shock response (HSR), causes extensive fat remodeling in peripheral tissues. These changes include a decrease in fat desaturase and acid lipase expression in the intestine and a global shift in the saturation levels of plasma membrane's phospholipids. The observed remodeling of plasma membrane is in line with ectothermic adaptive responses and gives worms a cumulative advantage to warm temperatures. We have determined that at least 6 TAX-2/TAX-4 cyclic guanosine monophosphate (cGMP) gated channel expressing sensory neurons, and transforming growth factor ß (TGF-β)/bone morphogenetic protein (BMP) are required for signaling across tissues to modulate fat desaturation. We also find neuronal hsf-1 is not only sufficient but also partially necessary to control the fat remodeling response and for survival at warm temperatures. This is the first study to show that a thermostat-based mechanism can cell nonautonomously coordinate membrane saturation and composition across tissues in a multicellular animal.
Collapse
Affiliation(s)
- Laetitia Chauve
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | - Francesca Hodge
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | - Sharlene Murdoch
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | - Greg West
- Babraham Institute, Cambridge, United Kingdom
| | | | | | - Cheryl Li
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | | | | | - Karin Kleigrewe
- Bavarian Centre for Biomolecular Mass Spectrometry, Freising, Germany
| | - Mario de Bono
- Institute of Science and Technology, Klosterneuburg, Austria
| | | | - Olivia Casanueva
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
15
|
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: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
16
|
Mørch MGM, Møller KV, Hesselager MO, Harders RH, Kidmose CL, Buhl T, Fuursted K, Bendixen E, Shen C, Christensen LG, Poulsen CH, Olsen A. The TGF-β ligand DBL-1 is a key player in a multifaceted probiotic protection against MRSA in C. elegans. Sci Rep 2021; 11:10717. [PMID: 34021197 PMCID: PMC8139972 DOI: 10.1038/s41598-021-89831-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023] Open
Abstract
Worldwide the increase in multi-resistant bacteria due to misuse of traditional antibiotics is a growing threat for our health. Finding alternatives to traditional antibiotics is thus timely. Probiotic bacteria have numerous beneficial effects and could offer safer alternatives to traditional antibiotics. Here, we use the nematode Caenorhabditis elegans (C. elegans) to screen a library of different lactobacilli to identify potential probiotic bacteria and characterize their mechanisms of action. We show that pretreatment with the Lactobacillus spp. Lb21 increases lifespan of C. elegans and results in resistance towards pathogenic methicillin-resistant Staphylococcus aureus (MRSA). Using genetic analysis, we find that Lb21-mediated MRSA resistance is dependent on the DBL-1 ligand of the TGF-β signaling pathway in C. elegans. This response is evolutionarily conserved as we find that Lb21 also induces the TGF-β pathway in porcine epithelial cells. We further characterize the host responses in an unbiased proteome analysis and identify 474 proteins regulated in worms fed Lb21 compared to control food. These include fatty acid CoA synthetase ACS-22, aspartic protease ASP-6 and vitellogenin VIT-2 which are important for Lb21-mediated MRSA resistance. Thus, Lb21 exerts its probiotic effect on C. elegans in a multifactorial manner. In summary, our study establishes a mechanistic basis for the antimicrobial potential of lactobacilli.
Collapse
Affiliation(s)
- Maria G M Mørch
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Katrine V Møller
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | | | - Rikke H Harders
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Caroline L Kidmose
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Therese Buhl
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | | | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Chong Shen
- Gut Immunology Lab, Health & Biosciences , IFF , Brabrand , Denmark
| | | | | | - Anders Olsen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
17
|
George AK, Behera J, Homme RP, Tyagi N, Tyagi SC, Singh M. Rebuilding Microbiome for Mitigating Traumatic Brain Injury: Importance of Restructuring the Gut-Microbiome-Brain Axis. Mol Neurobiol 2021; 58:3614-3627. [PMID: 33774742 PMCID: PMC8003896 DOI: 10.1007/s12035-021-02357-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/10/2021] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a damage to the brain from an external force that results in temporary or permanent impairment in brain functions. Unfortunately, not many treatment options are available to TBI patients. Therefore, knowledge of the complex interplay between gut microbiome (GM) and brain health may shed novel insights as it is a rapidly expanding field of research around the world. Recent studies show that GM plays important roles in shaping neurogenerative processes such as blood-brain-barrier (BBB), myelination, neurogenesis, and microglial maturation. In addition, GM is also known to modulate many aspects of neurological behavior and cognition; however, not much is known about the role of GM in brain injuries. Since GM has been shown to improve cellular and molecular functions via mitigating TBI-induced pathologies such as BBB permeability, neuroinflammation, astroglia activation, and mitochondrial dysfunction, herein we discuss how a dysbiotic gut environment, which in fact, contributes to central nervous system (CNS) disorders during brain injury and how to potentially ward off these harmful effects. We further opine that a better understanding of GM-brain (GMB) axis could help assist in designing better treatment and management strategies in future for the patients who are faced with limited options.
Collapse
Affiliation(s)
- Akash K George
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA
| | - Jyotirmaya Behera
- Bone Biology Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA
| | - Rubens P Homme
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA
| | - Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA. .,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, 40202, USA.
| |
Collapse
|
18
|
LRIG proteins regulate lipid metabolism via BMP signaling and affect the risk of type 2 diabetes. Commun Biol 2021; 4:90. [PMID: 33469151 PMCID: PMC7815736 DOI: 10.1038/s42003-020-01613-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeats and immunoglobulin-like domains (LRIG) proteins have been implicated as regulators of growth factor signaling; however, the possible redundancy among mammalian LRIG1, LRIG2, and LRIG3 has hindered detailed elucidation of their physiological functions. Here, we show that Lrig-null mouse embryonic fibroblasts (MEFs) are deficient in adipogenesis and bone morphogenetic protein (BMP) signaling. In contrast, transforming growth factor-beta (TGF-β) and receptor tyrosine kinase (RTK) signaling appeared unaltered in Lrig-null cells. The BMP signaling defect was rescued by ectopic expression of LRIG1 or LRIG3 but not by expression of LRIG2. Caenorhabditis elegans with mutant LRIG/sma-10 variants also exhibited a lipid storage defect. Human LRIG1 variants were strongly associated with increased body mass index (BMI) yet protected against type 2 diabetes; these effects were likely mediated by altered adipocyte morphology. These results demonstrate that LRIG proteins function as evolutionarily conserved regulators of lipid metabolism and BMP signaling and have implications for human disease.
Collapse
|
19
|
Bosch PJ, Peek SL, Smolikove S, Weiner JA. Akirin proteins in development and disease: critical roles and mechanisms of action. Cell Mol Life Sci 2020; 77:4237-4254. [PMID: 32361777 PMCID: PMC7606436 DOI: 10.1007/s00018-020-03531-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/05/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022]
Abstract
The Akirin genes, which encode small, nuclear proteins, were first characterized in 2008 in Drosophila and rodents. Early studies demonstrated important roles in immune responses and tumorigenesis, which subsequent work found to be highly conserved. More recently, a multiplicity of Akirin functions, and the associated molecular mechanisms involved, have been uncovered. Here, we comprehensively review what is known about invertebrate Akirin and its two vertebrate homologues Akirin1 and Akirin2, highlighting their role in regulating gene expression changes across a number of biological systems. We detail essential roles for Akirin family proteins in the development of the brain, limb, and muscle, in meiosis, and in tumorigenesis, emphasizing associated signaling pathways. We describe data supporting the hypothesis that Akirins act as a "bridge" between a variety of transcription factors and major chromatin remodeling complexes, and discuss several important questions remaining to be addressed. In little more than a decade, Akirin proteins have gone from being completely unknown to being increasingly recognized as evolutionarily conserved mediators of gene expression programs essential for the formation and function of animals.
Collapse
Affiliation(s)
- Peter J Bosch
- Department of Biology and Iowa Neuroscience Institute, University of Iowa, 143 Biology Building, Iowa City, IA, 52242, USA
| | - Stacey L Peek
- Interdisciplinary Graduate Program in Neuroscience, Department of Biology and Iowa Neuroscience Institute, University of Iowa, 143 Biology Building, Iowa City, IA, 52242, USA
| | - Sarit Smolikove
- Department of Biology, University of Iowa, 143 Biology Building, Iowa City, IA, 52242, USA
| | - Joshua A Weiner
- Department of Biology and Iowa Neuroscience Institute, University of Iowa, 143 Biology Building, Iowa City, IA, 52242, USA.
| |
Collapse
|
20
|
Abstract
Abstract
Background
Organisms show an incredibly diverse array of body and organ shapes that are both unique to their taxon and important for adapting to their environment. Achieving these specific shapes involves coordinating the many processes that transform single cells into complex organs, and regulating their growth so that they can function within a fully-formed body.
Main text
Conceptually, body and organ shape can be separated in two categories, although in practice these categories need not be mutually exclusive. Body shape results from the extent to which organs, or parts of organs, grow relative to each other. The patterns of relative organ size are characterized using allometry. Organ shape, on the other hand, is defined as the geometric features of an organ’s component parts excluding its size. Characterization of organ shape is frequently described by the relative position of homologous features, known as landmarks, distributed throughout the organ. These descriptions fall into the domain of geometric morphometrics.
Conclusion
In this review, we discuss the methods of characterizing body and organ shape, the developmental programs thought to underlie each, highlight when and how the mechanisms regulating body and organ shape might overlap, and provide our perspective on future avenues of research.
Collapse
|
21
|
Cohen JD, Sundaram MV. C. elegans Apical Extracellular Matrices Shape Epithelia. J Dev Biol 2020; 8:E23. [PMID: 33036165 PMCID: PMC7712855 DOI: 10.3390/jdb8040023] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Apical extracellular matrices (aECMs) coat exposed surfaces of epithelia to shape developing tissues and protect them from environmental insults. Despite their widespread importance for human health, aECMs are poorly understood compared to basal and stromal ECMs. The nematode Caenorhabditis elegans contains a variety of distinct aECMs, some of which share many of the same types of components (lipids, lipoproteins, collagens, zona pellucida domain proteins, chondroitin glycosaminoglycans and proteoglycans) with mammalian aECMs. These aECMs include the eggshell, a glycocalyx-like pre-cuticle, both collagenous and chitin-based cuticles, and other understudied aECMs of internal epithelia. C. elegans allows rapid genetic manipulations and live imaging of fluorescently-tagged aECM components, and is therefore providing new insights into aECM structure, trafficking, assembly, and functions in tissue shaping.
Collapse
Affiliation(s)
| | - Meera V. Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine 415 Curie Blvd, Philadelphia, PA 19104-6145, USA;
| |
Collapse
|
22
|
CREB Non-autonomously Controls Reproductive Aging through Hedgehog/Patched Signaling. Dev Cell 2020; 54:92-105.e5. [PMID: 32544391 DOI: 10.1016/j.devcel.2020.05.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/28/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022]
Abstract
Evolutionarily conserved signaling pathways are crucial for adjusting growth, reproduction, and cell maintenance in response to altered environmental conditions or energy balance. However, we have an incomplete understanding of the signaling networks and mechanistic changes that coordinate physiological changes across tissues. We found that loss of the cAMP response element-binding protein (CREB) transcription factor significantly slows Caenorhabditis elegans' reproductive decline, an early hallmark of aging in many animals. Our results indicate that CREB acts downstream of the transforming growth factor β (TGF-β) Sma/Mab pathway in the hypodermis to control reproductive aging, and that it does so by regulating a Hedgehog-related signaling factor, WRT-10. Overexpression of hypodermal wrt-10 is sufficient to delay reproductive decline and oocyte quality deterioration, potentially acting via Patched-related receptors in the germline. This TGF-β-CREB-Hedgehog signaling axis allows a key metabolic tissue to communicate with the reproductive system to regulate oocyte quality and the rate of reproductive decline.
Collapse
|
23
|
Abete-Luzi P, Fukushige T, Yun S, Krause MW, Eisenmann DM. New Roles for the Heterochronic Transcription Factor LIN-29 in Cuticle Maintenance and Lipid Metabolism at the Larval-to-Adult Transition in Caenorhabditis elegans. Genetics 2020; 214:669-690. [PMID: 31974205 PMCID: PMC7054012 DOI: 10.1534/genetics.119.302860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/21/2020] [Indexed: 11/18/2022] Open
Abstract
Temporal regulation of gene expression is a crucial aspect of metazoan development. In the roundworm Caenorhabditis elegans, the heterochronic pathway controls multiple developmental events in a time-specific manner. The most downstream effector of this pathway, the zinc-finger transcription factor LIN-29, acts in the last larval stage (L4) to regulate elements of the larval-to-adult switch. Here, we explore new LIN-29 targets and their implications for this developmental transition. We used RNA-sequencing to identify genes differentially expressed between animals misexpressing LIN-29 at an early time point and control animals. Among 230 LIN-29-activated genes, we found that genes encoding cuticle collagens were overrepresented. Interestingly, expression of lin-29 and some of these collagens was increased in adults with cuticle damage, suggesting a previously unknown function for LIN-29 in adult cuticle maintenance. On the other hand, genes involved in fat metabolism were enriched among 350 LIN-29-downregulated targets. We used mass spectrometry to assay lipid content in animals overexpressing LIN-29 and observed reduced fatty acid levels. Many LIN-29-repressed genes are normally expressed in the intestine, suggesting cell-nonautonomous regulation. We identified several LIN-29 upregulated genes encoding signaling molecules that may act as mediators in the regulation of intestinally expressed genes encoding fat metabolic enzymes and vitellogenins. Overall, our results support the model of LIN-29 as a major regulator of adult cuticle synthesis and integrity, and as the trigger for metabolic changes that take place at the important transition from rapid growth during larval life to slower growth and offspring production during adulthood.
Collapse
Affiliation(s)
- Patricia Abete-Luzi
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - Tetsunari Fukushige
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Sijung Yun
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael W Krause
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| |
Collapse
|
24
|
Akirin Is Required for Muscle Function and Acts Through the TGF-β Sma/Mab Signaling Pathway in Caenorhabditis elegans Development. G3-GENES GENOMES GENETICS 2020; 10:387-400. [PMID: 31767636 PMCID: PMC6945016 DOI: 10.1534/g3.119.400377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Akirin, a conserved metazoan protein, functions in muscle development in flies and mice. However, this was only tested in the rodent and fly model systems. Akirin was shown to act with chromatin remodeling complexes in transcription and was established as a downstream target of the NFκB pathway. Here we show a role for Caenorhabditis elegans Akirin/AKIR-1 in the muscle and body length regulation through a different pathway. Akirin localizes to somatic tissues throughout the body of C. elegans, including muscle nuclei. In agreement with its role in other model systems, Akirin loss of function mutants exhibit defects in muscle development in the embryo, as well as defects in movement and maintenance of muscle integrity in the C. elegans adult. We also have determined that Akirin acts downstream of the TGF-β Sma/Mab signaling pathway in controlling body size. Moreover, we found that the loss of Akirin resulted in an increase in autophagy markers, similar to mutants in the TGF-β Sma/Mab signaling pathway. In contrast to what is known in rodent and fly models, C. elegans Akirin does not act with the SWI/SNF chromatin-remodeling complex, and is instead involved with the NuRD chromatin remodeling complex in both movement and regulation of body size. Our studies define a novel developmental role (body size) and a new pathway (TGF-β Sma/Mab) for Akirin function, and confirmed its evolutionarily conserved function in muscle development in a new organism.
Collapse
|
25
|
Zheng S, Qu Z, Zanetti M, Lam B, Chin-Sang I. C. elegans PTEN and AMPK block neuroblast divisions by inhibiting a BMP-insulin-PP2A-MAPK pathway. Development 2018; 145:145/23/dev166876. [PMID: 30487179 DOI: 10.1242/dev.166876] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022]
Abstract
Caenorhabditis elegans that hatch in the absence of food stop their postembryonic development in a process called L1 arrest. Intriguingly, we find that the postembryonic Q neuroblasts divide and migrate during L1 arrest in mutants that have lost the energy sensor AMP-activated protein kinase (AMPK) or the insulin/IGF-1 signaling (IIS) negative regulator DAF-18/PTEN. We report that DBL-1/BMP works upstream of IIS to promote agonistic insulin-like peptides during L1 arrest. However, the abnormal Q cell divisions that occur during L1 arrest use a novel branch of the IIS pathway that is independent of the terminal transcription factor DAF-16/FOXO. Using genetic epistasis and drug interactions we show that AMPK functions downstream of, or in parallel with DAF-18/PTEN and IIS to inhibit PP2A function. Further, we show that PP2A regulates the abnormal Q cell divisions by activating the MPK-1/ERK signaling pathway via LIN-45/RAF, independently of LET-60/RAS. PP2A acts as a tumor suppressor in many oncogenic signaling cascades. Our work demonstrates a new role for PP2A that is needed to induce neuroblast divisions during starvation and is regulated by both insulin and AMPK.
Collapse
Affiliation(s)
- Shanqing Zheng
- Department of Biology, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Zhi Qu
- Department of Biology, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Michael Zanetti
- Department of Biology, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Brandon Lam
- Department of Biology, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Ian Chin-Sang
- Department of Biology, Queen's University, Kingston, ON, Canada K7L 3N6
| |
Collapse
|
26
|
BMP Signaling Determines Body Size via Transcriptional Regulation of Collagen Genes in Caenorhabditis elegans. Genetics 2018; 210:1355-1367. [PMID: 30274988 DOI: 10.1534/genetics.118.301631] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/25/2018] [Indexed: 01/10/2023] Open
Abstract
Body size is a tightly regulated phenotype in metazoans that depends on both intrinsic and extrinsic factors. While signaling pathways are known to control organ and body size, the downstream effectors that mediate their effects remain poorly understood. In the nematode Caenorhabditis elegans, a Bone Morphogenetic Protein (BMP)-related signaling pathway is the major regulator of growth and body size. We investigated the transcriptional network through which the BMP pathway regulates body size and identified cuticle collagen genes as major effectors of growth control. We demonstrate that cuticle collagens can act as positive regulators (col-41), negative regulators (col-141), or dose-sensitive regulators (rol-6) of body size. Moreover, we find a requirement of BMP signaling for stage-specific expression of cuticle collagen genes. We show that the Smad signal transducers directly bind conserved Smad-binding elements in regulatory regions of col-141 and col-142, but not of col-41 Hence, cuticle collagen genes may be directly and indirectly regulated via the BMP pathway. Our work thus connects a conserved signaling pathway with its critical downstream effectors, advancing insight into how body size is specified. Since collagen mutations and misregulation are implicated in numerous human genetic disorders and injury sequelae, understanding how collagen gene expression is regulated has broad implications.
Collapse
|
27
|
Clark JF, Savage-Dunn C. Delta-9 fatty acid desaturase mutants display increased body size. MICROPUBLICATION BIOLOGY 2018; 2018:10.17912/SS8E-6587. [PMID: 32550399 PMCID: PMC7282526 DOI: 10.17912/ss8e-6587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- James F. Clark
- Department of Biology, Queens College, CUNY 65-30 Kissena Boulevard Flushing, NY 11367,
Ph.D. Program in Biology, The Graduate Center, CUNY 365 5th Avenue New York, NY 10016,
Correspondence to: James F. Clark ()
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY 65-30 Kissena Boulevard Flushing, NY 11367,
Ph.D. Program in Biology, The Graduate Center, CUNY 365 5th Avenue New York, NY 10016
| |
Collapse
|
28
|
Membrane Fluidity Is Regulated Cell Nonautonomously by Caenorhabditis elegans PAQR-2 and Its Mammalian Homolog AdipoR2. Genetics 2018; 210:189-201. [PMID: 29997234 PMCID: PMC6116961 DOI: 10.1534/genetics.118.301272] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/09/2018] [Indexed: 01/24/2023] Open
Abstract
The properties of cell membranes are determined mostly by the types of fatty acids that they contain. Bodhicharla et al. report that a key regulator of membrane fluidity, the PAQR-2/IGLR-2 protein complex... Maintenance of membrane properties is an essential aspect of cellular homeostasis of which the regulatory mechanisms remain mostly uncharacterized. In Caenorhabditis elegans, the PAQR-2 and IGLR-2 proteins act together as a plasma membrane sensor that responds to decreased fluidity by promoting fatty acid desaturation, hence restoring membrane fluidity. Here, we used mosaic analysis for paqr-2 and iglr-2, and tissue-specific paqr-2 expression, to show that membrane homeostasis is achieved cell nonautonomously. Specifically, we found that expression of paqr-2 in the hypodermis, gonad sheath cells, or intestine is sufficient to suppress systemic paqr-2 mutant phenotypes, including tail tip morphology, membrane fluidity in intestinal cells, cold and glucose intolerance, vitellogenin transport to the germline, germ cell development, and brood size. Finally, we show that the cell nonautonomous regulation of membrane homeostasis is conserved in human cells: HEK293 cells that express AdipoR2, a homolog of paqr-2, are able to normalize membrane fluidity in distant cells where AdipoR2 has been silenced. Finally, using C. elegans mutants and small interfering RNA against Δ9 stearoyl-CoA desaturase in HEK293 cells, we show that Δ9 desaturases are essential for the cell nonautonomous maintenance of membrane fluidity. We conclude that cells are able to share membrane components even when they are not in direct contact with each other, and that this contributes to the maintenance of membrane homeostasis in C. elegans and human cells.
Collapse
|