1
|
Liu X, Chang Z, Sun P, Cao B, Wang Y, Fang J, Pei Y, Chen B, Zou W. MONITTR allows real-time imaging of transcription and endogenous proteins in C. elegans. J Cell Biol 2025; 224:e202403198. [PMID: 39400293 PMCID: PMC11473600 DOI: 10.1083/jcb.202403198] [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: 04/01/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Maximizing cell survival under stress requires rapid and transient adjustments of RNA and protein synthesis. However, capturing these dynamic changes at both single-cell level and across an organism has been challenging. Here, we developed a system named MONITTR (MS2-embedded mCherry-based monitoring of transcription) for real-time simultaneous measurement of nascent transcripts and endogenous protein levels in C. elegans. Utilizing this system, we monitored the transcriptional bursting of fasting-induced genes and found that the epidermis responds to fasting by modulating the proportion of actively transcribing nuclei and transcriptional kinetics of individual alleles. Additionally, our findings revealed the essential roles of the transcription factors NHR-49 and HLH-30 in governing the transcriptional kinetics of fasting-induced genes under fasting. Furthermore, we tracked transcriptional dynamics during heat-shock response and ER unfolded protein response and observed rapid changes in the level of nascent transcripts under stress conditions. Collectively, our study provides a foundation for quantitatively investigating how animals spatiotemporally modulate transcription in various physiological and pathological conditions.
Collapse
Affiliation(s)
- Xiaofan Liu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Chang
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Pingping Sun
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Beibei Cao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Yuzhi Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jie Fang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- Department of Cell Biology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yechun Pei
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Baohui Chen
- Department of Cell Biology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Al-Refaie N, Padovani F, Hornung J, Pudelko L, Binando F, Del Carmen Fabregat A, Zhao Q, Towbin BD, Cenik ES, Stroustrup N, Padeken J, Schmoller KM, Cabianca DS. Fasting shapes chromatin architecture through an mTOR/RNA Pol I axis. Nat Cell Biol 2024:10.1038/s41556-024-01512-w. [PMID: 39300311 DOI: 10.1038/s41556-024-01512-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Chromatin architecture is a fundamental mediator of genome function. Fasting is a major environmental cue across the animal kingdom, yet how it impacts three-dimensional (3D) genome organization is unknown. Here we show that fasting induces an intestine-specific, reversible and large-scale spatial reorganization of chromatin in Caenorhabditis elegans. This fasting-induced 3D genome reorganization requires inhibition of the nutrient-sensing mTOR pathway, acting through the regulation of RNA Pol I, but not Pol II nor Pol III, and is accompanied by remodelling of the nucleolus. By uncoupling the 3D genome configuration from the animal's nutritional status, we find that the expression of metabolic and stress-related genes increases when the spatial reorganization of chromatin occurs, showing that the 3D genome might support the transcriptional response in fasted animals. Our work documents a large-scale chromatin reorganization triggered by fasting and reveals that mTOR and RNA Pol I shape genome architecture in response to nutrients.
Collapse
Affiliation(s)
- Nada Al-Refaie
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians Universität München, Munich, Germany
| | - Francesco Padovani
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johanna Hornung
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lorenz Pudelko
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians Universität München, Munich, Germany
| | - Francesca Binando
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Andrea Del Carmen Fabregat
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Qiuxia Zhao
- Department of Molecular Biosciences, University of Texas Austin, Austin, TX, USA
| | | | - Elif Sarinay Cenik
- Department of Molecular Biosciences, University of Texas Austin, Austin, TX, USA
| | - Nicholas Stroustrup
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jan Padeken
- Institute of Molecular Biology, Mainz, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Daphne S Cabianca
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
3
|
Du J, Liu W, Li M, Li Z, Li X, Dai Y, Liu G, Wang X, Zhu P, Gladyshev VN, Zhou X. Comparative time-series multi-omics analyses suggest H1.2 involvement in anoxic adaptation and cancer resistance. PLoS Biol 2024; 22:e3002778. [PMID: 39178313 PMCID: PMC11376556 DOI: 10.1371/journal.pbio.3002778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/05/2024] [Accepted: 07/31/2024] [Indexed: 08/25/2024] Open
Abstract
The naked mole rat (NMR), Heterocephalus glaber, is known as the longest-lived rodent and is extraordinarily resistant to hypoxia and cancer. Here, both NMR embryonic fibroblasts (NEFs) and their mouse counterparts (MEFs) were subjected to anoxic conditions (0% O2, 5% CO2). A combination of comparative transcriptomics and proteomics was then employed to identify differentially expressed genes (DEGs). Notably, we observed distinct levels of histone H1.2 (encoded by HIST1H1C) accumulation between NEFs and MEFs. Subsequent mechanistic analyses showed that higher H1.2 expression in NEFs was associated with the lower expression of its inhibitor, PARP1. Additionally, we discovered that H1.2 can directly interact with HIF-1α PAS domains, thereby promoting the expression of HIF-1α through facilitating the dimerization with HIF-1β. The overexpression of H1.2 was also found to trigger autophagy and to suppress the migration of cancer cells, as well as the formation of xenograft tumors, via the NRF2/P62 signaling pathway. Moreover, an engineered H1.2 knock-in mouse model exhibited significantly extended survival in hypoxic conditions (4% O2) and showed a reduced rate of tumor formation. Collectively, our results indicate a potential mechanistic link between H1.2 and the dual phenomena of anoxic adaptation and cancer resistance.
Collapse
Affiliation(s)
- Juan Du
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weiqiang Liu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| | - Zihao Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuanjing Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yichen Dai
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| | - Gaoming Liu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| | - Xiao Wang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| | - Pingfen Zhu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xuming Zhou
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chaoyang District, Beijing, China
| |
Collapse
|
4
|
Tan CH, Wang TY, Park H, Lomenick B, Chou TF, Sternberg PW. Single-tissue proteomics in Caenorhabditis elegans reveals proteins resident in intestinal lysosome-related organelles. Proc Natl Acad Sci U S A 2024; 121:e2322588121. [PMID: 38861598 PMCID: PMC11194598 DOI: 10.1073/pnas.2322588121] [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: 12/25/2023] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
The nematode intestine is the primary site for nutrient uptake and storage as well as the synthesis of biomolecules; lysosome-related organelles known as gut granules are important for many of these functions. Aspects of intestine biology are not well understood, including the export of the nutrients it imports and the molecules it synthesizes, as well as the complete functions and protein content of the gut granules. Here, we report a mass spectrometry (MS)-based proteomic analysis of the intestine of the Caenorhabditis elegans and of its gut granules. Overall, we identified approximately 5,000 proteins each in the intestine and the gonad and showed that most of these proteins can be detected in samples extracted from a single worm, suggesting the feasibility of individual-level genetic analysis using proteomes. Comparing proteomes and published transcriptomes of the intestine and the gonad, we identified proteins that appear to be synthesized in the intestine and then transferred to the gonad. To identify gut granule proteins, we compared the proteome of individual intestines deficient in gut granules to the wild type. The identified gut granule proteome includes proteins known to be exclusively localized to the granules and additional putative gut granule proteins. We selected two of these putative gut granule proteins for validation via immunohistochemistry, and our successful confirmation of both suggests that our strategy was effective in identifying the gut granule proteome. Our results demonstrate the practicability of single-tissue MS-based proteomic analysis in small organisms and in its future utility.
Collapse
Affiliation(s)
- Chieh-Hsiang Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Ting-Yu Wang
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| | - Heenam Park
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
5
|
Chen TY, Wang F, Lee P, Hsu A, Ching T. Mitochondrial S-adenosylmethionine deficiency induces mitochondrial unfolded protein response and extends lifespan in Caenorhabditis elegans. Aging Cell 2024; 23:e14103. [PMID: 38361361 PMCID: PMC11019128 DOI: 10.1111/acel.14103] [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: 05/05/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
S-adenosylmethionine (SAM), generated from methionine and ATP by S-adenosyl methionine synthetase (SAMS), is the universal methyl group donor required for numerous cellular methylation reactions. In Caenorhabditis elegans, silencing sams-1, the major isoform of SAMS, genetically or via dietary restriction induces a robust mitochondrial unfolded protein response (UPRmt) and lifespan extension. In this study, we found that depleting SAMS-1 markedly decreases mitochondrial SAM levels. Moreover, RNAi knockdown of SLC-25A26, a carrier protein responsible for transporting SAM from the cytoplasm into the mitochondria, significantly lowers the mitochondrial SAM levels and activates UPRmt, suggesting that the UPRmt induced by sams-1 mutations might result from disrupted mitochondrial SAM homeostasis. Through a genetic screen, we then identified a putative mitochondrial tRNA methyltransferase TRMT-10C.2 as a major downstream effector of SAMS-1 to regulate UPRmt and longevity. As disruption of mitochondrial tRNA methylation likely leads to impaired mitochondrial tRNA maturation and consequently reduced mitochondrial translation, our findings suggest that depleting mitochondrial SAM level might trigger UPRmt via attenuating protein translation in the mitochondria. Together, this study has revealed a potential mechanism by which SAMS-1 regulates UPRmt and longevity.
Collapse
Affiliation(s)
- Tse Yu Chen
- Institute of Biopharmaceutical SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Feng‐Yung Wang
- Institute of Biochemistry and Molecular BiologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Pin‐Jung Lee
- Institute of Biopharmaceutical SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ao‐Lin Hsu
- Institute of Biochemistry and Molecular BiologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Biological Science & Technology and Institute of Biochemistry and Molecular BiologyChina Medical UniversityTaichungTaiwan
- Department of Internal Medicine, Division of Geriatric and Palliative MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Tsui‐Ting Ching
- Institute of Biopharmaceutical SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
6
|
Theska T, Renahan T, Sommer RJ. Starvation resistance in the nematode Pristionchus pacificus requires a conserved supplementary nuclear receptor. ZOOLOGICAL LETTERS 2024; 10:7. [PMID: 38481284 PMCID: PMC10938818 DOI: 10.1186/s40851-024-00227-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/18/2024] [Indexed: 03/17/2024]
Abstract
Nuclear hormone receptors (NHRs) are a deeply-conserved superfamily of metazoan transcription factors, which fine-tune the expression of their regulatory target genes in response to a plethora of sensory inputs. In nematodes, NHRs underwent an explosive expansion and many species have hundreds of nhr genes, most of which remain functionally uncharacterized. However, recent studies have reported that two sister receptors, Ppa-NHR-1 and Ppa-NHR-40, are crucial regulators of feeding-structure morphogenesis in the diplogastrid model nematode Pristionchus pacificus. In the present study, we functionally characterize Ppa-NHR-10, the sister paralog of Ppa-NHR-1 and Ppa-NHR-40, aiming to reveal whether it too regulates aspects of feeding-structure development. We used CRISPR/CAS9-mediated mutagenesis to create small frameshift mutations of this nuclear receptor gene and applied a combination of geometric morphometrics and unsupervised clustering to characterize potential mutant phenotypes. However, we found that Ppa-nhr-10 mutants do not show aberrant feeding-structure morphologies. Instead, multiple RNA-seq experiments revealed that many of the target genes of this receptor are involved in lipid catabolic processes. We hypothesized that their mis-regulation could affect the survival of mutant worms during starvation, where lipid catabolism is often essential. Indeed, using novel survival assays, we found that mutant worms show drastically decreased starvation resistance, both as young adults and as dauer larvae. We also characterized genome-wide changes to the transcriptional landscape in P. pacificus when exposed to 24 h of acute starvation, and found that Ppa-NHR-10 partially regulates some of these responses. Taken together, these results demonstrate that Ppa-NHR-10 is broadly required for starvation resistance and regulates different biological processes than its closest paralogs Ppa-NHR-1 and Ppa-NHR-40.
Collapse
Affiliation(s)
- Tobias Theska
- Department for Integrative Evolutionary Biology, Max Planck Institute for Biology, Tübingen, Max-Planck-Ring 9, 72076, Tübingen, Germany
| | - Tess Renahan
- Department for Integrative Evolutionary Biology, Max Planck Institute for Biology, Tübingen, Max-Planck-Ring 9, 72076, Tübingen, Germany
| | - Ralf J Sommer
- Department for Integrative Evolutionary Biology, Max Planck Institute for Biology, Tübingen, Max-Planck-Ring 9, 72076, Tübingen, Germany.
| |
Collapse
|
7
|
Yang J, Liu W, Han X, Hao X, Yao Q, Du W. Gut microbiota modulation enhances the immune capacity of lizards under climate warming. MICROBIOME 2024; 12:37. [PMID: 38388458 PMCID: PMC10882899 DOI: 10.1186/s40168-023-01736-2] [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: 09/27/2023] [Accepted: 12/04/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND Host-microbial interactions are expected to affect species' adaptability to climate change but have rarely been explored in ectothermic animals. Some studies have shown that short-term warming reduced gut microbial diversity that could hamper host functional performance. RESULTS However, our longitudinal experiments in semi-natural conditions demonstrated that warming decreased gut microbiota diversity at 2 months, but increased diversity at 13 and 27 months in a desert lizard (Eremias multiocellata). Simultaneously, long-term warming significantly increased the antibacterial activity of serum, immune responses (higher expression of intestinal immune-related genes), and the concentration of short-chain fatty acids (thereby intestinal barrier and immunity) in the lizard. Fecal microbiota transplant experiments further revealed that increased diversity of gut microbiota significantly enhanced antibacterial activity and the immune response of lizards. More specifically, the enhanced immunity is likely due to the higher relative abundance of Bacteroides in warming lizards, given that the bacteria of Bacteroides fragilis regulated IFN-β expression to increase the immune response of lizards under a warming climate. CONCLUSIONS Our study suggests that gut microbiota can help ectotherms cope with climate warming by enhancing host immune response, and highlights the importance of long-term studies on host-microbial interactions and their biological impacts.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiqiang Liu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xingzhi Han
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Wildlife and Protected Areas, Northeast Forestry University, Harbin, 150040, China
| | - Xin Hao
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural, School of Rural Revitalization), Hainan University, Danzhou, 571737, China
| | - Qibin Yao
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiguo Du
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
8
|
Doering KRS, Ermakova G, Taubert S. Nuclear hormone receptor NHR-49 is an essential regulator of stress resilience and healthy aging in Caenorhabditis elegans. Front Physiol 2023; 14:1241591. [PMID: 37645565 PMCID: PMC10461480 DOI: 10.3389/fphys.2023.1241591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023] Open
Abstract
The genome of Caenorhabditis elegans encodes 284 nuclear hormone receptor, which perform diverse functions in development and physiology. One of the best characterized of these is NHR-49, related in sequence and function to mammalian hepatocyte nuclear factor 4α and peroxisome proliferator-activated receptor α. Initially identified as regulator of lipid metabolism, including fatty acid catabolism and desaturation, additional important roles for NHR-49 have since emerged. It is an essential contributor to longevity in several genetic and environmental contexts, and also plays vital roles in the resistance to several stresses and innate immune response to infection with various bacterial pathogens. Here, we review how NHR-49 is integrated into pertinent signaling circuits and how it achieves its diverse functions. We also highlight areas for future investigation including identification of regulatory inputs that drive NHR-49 activity and identification of tissue-specific gene regulatory outputs. We anticipate that future work on this protein will provide information that could be useful for developing strategies to age-associated declines in health and age-related human diseases.
Collapse
Affiliation(s)
- Kelsie R. S. Doering
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Glafira Ermakova
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
9
|
Scott A, Willis CR, Muratani M, Higashitani A, Etheridge T, Szewczyk NJ, Deane CS. Caenorhabditis elegans in microgravity: An omics perspective. iScience 2023; 26:107189. [PMID: 37456835 PMCID: PMC10344948 DOI: 10.1016/j.isci.2023.107189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The application of omics to study Caenorhabditis elegans (C. elegans) in the context of spaceflight is increasing, illuminating the wide-ranging biological impacts of spaceflight on physiology. In this review, we highlight the application of omics, including transcriptomics, genomics, proteomics, multi-omics, and integrated omics in the study of spaceflown C. elegans, and discuss the impact, use, and future direction of this branch of research. We highlight the variety of molecular alterations that occur in response to spaceflight, most notably changes in metabolic and neuromuscular gene regulation. These transcriptional features are reproducible and evident across many spaceflown species (e.g., mice and astronauts), supporting the use of C. elegans as a model organism to study spaceflight physiology with translational capital. Integrating tissue-specific, spatial, and multi-omics approaches, which quantitatively link molecular responses to phenotypic adaptations, will facilitate the identification of candidate regulatory molecules for therapeutic intervention and thus represents the next frontiers in C. elegans space omics research.
Collapse
Affiliation(s)
- Amanda Scott
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Craig R.G. Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Masafumi Muratani
- Transborder Medical Research Center and Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | - Timothy Etheridge
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Nathaniel J. Szewczyk
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Ohio Musculoskeletal and Neurological Institute, Ohio University, Athens, OH, USA
| | - Colleen S. Deane
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
10
|
Ghaddar A, Armingol E, Huynh C, Gevirtzman L, Lewis NE, Waterston R, O’Rourke EJ. Whole-body gene expression atlas of an adult metazoan. SCIENCE ADVANCES 2023; 9:eadg0506. [PMID: 37352352 PMCID: PMC10289653 DOI: 10.1126/sciadv.adg0506] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Gene activity defines cell identity, drives intercellular communication, and underlies the functioning of multicellular organisms. We present the single-cell resolution atlas of gene activity of a fertile adult metazoan: Caenorhabditis elegans. This compendium comprises 180 distinct cell types and 19,657 expressed genes. We predict 7541 transcription factor expression profile associations likely responsible for defining cellular identity. We predict thousands of intercellular interactions across the C. elegans body and the ligand-receptor pairs that mediate them, some of which we experimentally validate. We identify 172 genes that show consistent expression across cell types, are involved in basic and essential functions, and are conserved across phyla; therefore, we present them as experimentally validated housekeeping genes. We developed the WormSeq application to explore these data. In addition to the integrated gene-to-systems biology, we present genome-scale single-cell resolution testable hypotheses that we anticipate will advance our understanding of the molecular mechanisms, underlying the functioning of a multicellular organism and the perturbations that lead to its malfunction.
Collapse
Affiliation(s)
- Abbas Ghaddar
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Erick Armingol
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chau Huynh
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Louis Gevirtzman
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Nathan E. Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert Waterston
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Eyleen J. O’Rourke
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
11
|
Ghaddar A, Mony VK, Mishra S, Berhanu S, Johnson JC, Enriquez-Hesles E, Harrison E, Patel A, Horak MK, Smith JS, O'Rourke EJ. Increased alcohol dehydrogenase 1 activity promotes longevity. Curr Biol 2023; 33:1036-1046.e6. [PMID: 36805847 PMCID: PMC10236445 DOI: 10.1016/j.cub.2023.01.059] [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: 08/05/2022] [Revised: 11/28/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023]
Abstract
Several molecules can extend healthspan and lifespan across organisms. However, most are upstream signaling hubs or transcription factors orchestrating complex anti-aging programs. Therefore, these molecules point to but do not reveal the fundamental mechanisms driving longevity. Instead, downstream effectors that are necessary and sufficient to promote longevity across conditions or organisms may reveal the fundamental anti-aging drivers. Toward this goal, we searched for effectors acting downstream of the transcription factor EB (TFEB), known as HLH-30 in C. elegans, because TFEB/HLH-30 is necessary across anti-aging interventions and its overexpression is sufficient to extend C. elegans lifespan and reduce biomarkers of aging in mammals including humans. As a result, we present an alcohol-dehydrogenase-mediated anti-aging response (AMAR) that is essential for C. elegans longevity driven by HLH-30 overexpression, caloric restriction, mTOR inhibition, and insulin-signaling deficiency. The sole overexpression of ADH-1 is sufficient to activate AMAR, which extends healthspan and lifespan by reducing the levels of glycerol-an age-associated and aging-promoting alcohol. Adh1 overexpression is also sufficient to promote longevity in yeast, and adh-1 orthologs are induced in calorically restricted mice and humans, hinting at ADH-1 acting as an anti-aging effector across phyla.
Collapse
Affiliation(s)
- Abbas Ghaddar
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Vinod K Mony
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Swarup Mishra
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Samuel Berhanu
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - James C Johnson
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Elisa Enriquez-Hesles
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Emma Harrison
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Aaroh Patel
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Mary Kate Horak
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Jeffrey S Smith
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Eyleen J O'Rourke
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA; Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
12
|
Wong SQ, Ryan CJ, Bonal DM, Mills J, Lapierre LR. Neuronal HLH-30/TFEB modulates peripheral mitochondrial fragmentation to improve thermoresistance in Caenorhabditis elegans. Aging Cell 2023; 22:e13741. [PMID: 36419219 PMCID: PMC10014052 DOI: 10.1111/acel.13741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 09/29/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Transcription factor EB (TFEB) is a conserved master transcriptional activator of autophagy and lysosomal genes that modulates organismal lifespan regulation and stress resistance. As neurons can coordinate organism-wide processes, we investigated the role of neuronal TFEB in stress resistance and longevity. To this end, the Caenorhabditis elegans TFEB ortholog, hlh-30, was rescued panneuronally in hlh-30 loss of function mutants. While important in the long lifespan of daf-2 animals, neuronal HLH-30/TFEB was not sufficient to restore normal lifespan in short-lived hlh-30 mutants. However, neuronal HLH-30/TFEB rescue mediated robust improvements in the heat stress resistance of wildtype but not daf-2 animals. Notably, these mechanisms can be uncoupled, as neuronal HLH-30/TFEB requires DAF-16/FOXO to regulate longevity but not thermoresistance. Through further transcriptomics profiling and functional analysis, we discovered that neuronal HLH-30/TFEB modulates neurotransmission through the hitherto uncharacterized protein W06A11.1 by inducing peripheral mitochondrial fragmentation and organismal heat stress resistance in a non-cell autonomous manner. Taken together, this study uncovers a novel mechanism of heat stress protection mediated by neuronal HLH-30/TFEB.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
| | - Catherine J. Ryan
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
| | - Dennis M. Bonal
- Pathobiology Graduate Program, Division of Biology & MedicineBrown UniversityProvidenceRhode IslandUSA
| | - Joslyn Mills
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
- Department of BiologyWheaton CollegeNortonMassachusettsUSA
| | - Louis R. Lapierre
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
- Département de Chimie et BiochimieUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Center for Precision MedicineMonctonNew BrunswickCanada
| |
Collapse
|
13
|
Woodhouse RM, Frolows N, Wang G, Hawdon A, Wong EHK, Dansereau LC, Su Y, Adair LD, New EJ, Philp AM, Tan WK, Philp A, Ashe A. Mitochondrial succinate dehydrogenase function is essential for sperm motility and male fertility. iScience 2022; 25:105573. [PMID: 36465130 PMCID: PMC9709242 DOI: 10.1016/j.isci.2022.105573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial health is crucial to sperm quality and male fertility, but the precise role of mitochondria in sperm function remains unclear. SDHA is a component of the succinate dehydrogenase (SDH) complex and plays a critical role in mitochondria. In humans, SDH activity is positively correlated with sperm quality, and mutations in SDHA are associated with Leigh Syndrome. Here we report that the C. elegans SDHA orthologue SDHA-2 is essential for male fertility: sdha-2 mutants produce dramatically fewer offspring due to defective sperm activation and motility, have hyperfused sperm mitochondria, and disrupted redox balance. Similar sperm motility defects in sdha-1 and icl-1 mutant animals suggest an imbalance in metabolites may underlie the fertility defect. Our results demonstrate a role for SDHA-2 in sperm motility and male reproductive health and establish an animal model of SDH deficiency-associated infertility.
Collapse
Affiliation(s)
- Rachel M. Woodhouse
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
- Division of Genome Science and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Natalya Frolows
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
- CSIRO Health and Biosecurity, Sydney, NSW 2113, Australia
| | - Guoqiang Wang
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Azelle Hawdon
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Edmund Heng Kin Wong
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
| | - Linda C. Dansereau
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent’s Clinical School, UNSW Medicine, University of NSW, Sydney, NSW 2010, Australia
| | - Yingying Su
- Sydney Microscopy and Microanalysis, The University of Sydney, Sydney, NSW 2006, Australia
| | - Liam D. Adair
- The University of Sydney, School of Chemistry, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Elizabeth J. New
- The University of Sydney, School of Chemistry, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ashleigh M. Philp
- St Vincent’s Clinical School, UNSW Medicine, University of NSW, Sydney, NSW 2010, Australia
| | - Wei Kang Tan
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
| | - Andrew Philp
- Centre for Healthy Ageing, Centenary Institute, Missenden Road, Sydney, NSW 2050, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Alyson Ashe
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Wang T, Li TC, Miao YH, Wu LN, Chen YQ, Huang DW, Xiao JH. The gender-specific impact of starvation on mitotypes diversity in adults of Drosophila melanogaster. Open Biol 2022; 12:220108. [PMID: 36167086 PMCID: PMC9514890 DOI: 10.1098/rsob.220108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In animals, starvation can increase the level of reactive oxygen species (ROS) in some tissues. Mitochondrial DNA (mtDNA) is more vulnerable to being attacked by ROS due to the lack of histone protection, leading to oxidative damage. However, whether starvation is associated with the genetic diversity of mtDNA remains unclear. Here, by using adult individuals of Drosophila melanogaster under three different feeding treatments (starvation, with the provision of only water, and normal feeding), based on the high-throughput sequencing results of the PCR amplicons of the partial sequences of the mitochondrial gene cytochrome c oxidase subunit I (mt-cox1), no significant difference in the mean number of mitochondrial haplotypes and the mean genetic distance of haplotypes within individuals were identified between the three treatment groups. Coupled with the low proportion of heterogeneous mt-cox1 sequences within each individual, it suggested that starvation had a limited impact on mitotype genetic diversity and mitochondrial function. Nevertheless, starvation could significantly increase the sequence number of haplotypes containing specific mutations, and for males with higher levels of mitochondrial heteroplasmy than females in the normal feeding group, starvation could further increase their mitochondrial heteroplasmy.
Collapse
Affiliation(s)
- Tao Wang
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Tian-Chu Li
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Yun-Heng Miao
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Luo-Nan Wu
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Yu-Qiao Chen
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Da-Wei Huang
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Jin-Hua Xiao
- College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
15
|
McLachlan IG, Kramer TS, Dua M, DiLoreto EM, Gomes MA, Dag U, Srinivasan J, Flavell SW. Diverse states and stimuli tune olfactory receptor expression levels to modulate food-seeking behavior. eLife 2022; 11:e79557. [PMID: 36044259 PMCID: PMC9433090 DOI: 10.7554/elife.79557] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022] Open
Abstract
Animals must weigh competing needs and states to generate adaptive behavioral responses to the environment. Sensorimotor circuits are thus tasked with integrating diverse external and internal cues relevant to these needs to generate context-appropriate behaviors. However, the mechanisms that underlie this integration are largely unknown. Here, we show that a wide range of states and stimuli converge upon a single Caenorhabditis elegans olfactory neuron to modulate food-seeking behavior. Using an unbiased ribotagging approach, we find that the expression of olfactory receptor genes in the AWA olfactory neuron is influenced by a wide array of states and stimuli, including feeding state, physiological stress, and recent sensory cues. We identify odorants that activate these state-dependent olfactory receptors and show that altered expression of these receptors influences food-seeking and foraging. Further, we dissect the molecular and neural circuit pathways through which external sensory information and internal nutritional state are integrated by AWA. This reveals a modular organization in which sensory and state-related signals arising from different cell types in the body converge on AWA and independently control chemoreceptor expression. The synthesis of these signals by AWA allows animals to generate sensorimotor responses that reflect the animal's overall state. Our findings suggest a general model in which sensory- and state-dependent transcriptional changes at the sensory periphery modulate animals' sensorimotor responses to meet their ongoing needs and states.
Collapse
Affiliation(s)
- Ian G McLachlan
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Talya S Kramer
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
- MIT Biology Graduate Program, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Malvika Dua
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Elizabeth M DiLoreto
- Department of Biology and Biotechnology, Worcester Polytechnic InstituteWorcesterUnited States
| | - Matthew A Gomes
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Ugur Dag
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Jagan Srinivasan
- Department of Biology and Biotechnology, Worcester Polytechnic InstituteWorcesterUnited States
| | - Steven W Flavell
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
16
|
Higgins DP, Weisman CM, Lui DS, D'Agostino FA, Walker AK. Defining characteristics and conservation of poorly annotated genes in Caenorhabditis elegans using WormCat 2.0. Genetics 2022; 221:6588682. [PMID: 35587742 PMCID: PMC9339291 DOI: 10.1093/genetics/iyac085] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/04/2022] [Indexed: 12/03/2022] Open
Abstract
Omics tools provide broad datasets for biological discovery. However, the computational tools for identifying important genes or pathways in RNA-seq, proteomics, or GWAS (Genome-Wide Association Study) data depend on Gene Ontogeny annotations and are biased toward well-described pathways. This limits their utility as poorly annotated genes, which could have novel functions, are often passed over. Recently, we developed an annotation and category enrichment tool for Caenorhabditis elegans genomic data, WormCat, which provides an intuitive visualization output. Unlike Gene Ontogeny-based enrichment tools, which exclude genes with no annotation information, WormCat 2.0 retains these genes as a special UNASSIGNED category. Here, we show that the UNASSIGNED gene category enrichment exhibits tissue-specific expression patterns and can include genes with biological functions identified in published datasets. Poorly annotated genes are often considered to be potentially species-specific and thus, of reduced interest to the biomedical community. Instead, we find that around 3% of the UNASSIGNED genes have human orthologs, including some linked to human diseases. These human orthologs themselves have little annotation information. A recently developed method that incorporates lineage relationships (abSENSE) indicates that the failure of BLAST to detect homology explains the apparent lineage specificity for many UNASSIGNED genes. This suggests that a larger subset could be related to human genes. WormCat provides an annotation strategy that allows the association of UNASSIGNED genes with specific phenotypes and known pathways. Building these associations in C. elegans, with its robust genetic tools, provides a path to further functional study and insight into these understudied genes.
Collapse
Affiliation(s)
- Daniel P Higgins
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester MA 01605, USA
| | - Caroline M Weisman
- Lewis-Sigler Institute for Quantitative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Dominique S Lui
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester MA 01605, USA
| | - Frank A D'Agostino
- Department of Applied Mathematics, Harvard University, Cambridge MA 02138, USA
| | - Amy K Walker
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester MA 01605, USA
| |
Collapse
|
17
|
Piazzesi A, Wang Y, Jackson J, Wischhof L, Zeisler-Diehl V, Scifo E, Oganezova I, Hoffmann T, Gómez Martín P, Bertan F, Wrobel CJJ, Schroeder FC, Ehninger D, Händler K, Schultze JL, Schreiber L, van Echten-Deckert G, Nicotera P, Bano D. CEST-2.2 overexpression alters lipid metabolism and extends longevity of mitochondrial mutants. EMBO Rep 2022; 23:e52606. [PMID: 35297148 PMCID: PMC9066074 DOI: 10.15252/embr.202152606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short‐lived mitochondrial mutants into long‐lived ones. We find that CEST‐2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest‐2.2 expression has a minor lifespan impact on wild‐type nematodes, whereas its overexpression markedly extends the lifespan of complex I‐deficient gas‐1(fc21) mutants. We profile the transcriptome and lipidome of cest‐2.2 overexpressing animals and show that CEST‐2.2 stimulates lipid metabolism and fatty acid beta‐oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET‐721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue‐specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.
Collapse
Affiliation(s)
- Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Yiru Wang
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ina Oganezova
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thorben Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Chester J J Wrobel
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kristian Händler
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany.,Department for Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Lukas Schreiber
- Institute of Cellular and Molecular Botany (IZMB), University of Bonn, Bonn, Germany
| | | | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
18
|
Yu Y, Guo S, Ren Y, Zhang J, Li M, Tian S, Wang J, Sun H, Zuo Y, Chen Y, Gong G, Zhang H, Xu Y. Quantitative Transcriptomic and Proteomic Analysis of Fruit Development and Ripening in Watermelon ( Citrullus lanatus). FRONTIERS IN PLANT SCIENCE 2022; 13:818392. [PMID: 35392508 PMCID: PMC8980866 DOI: 10.3389/fpls.2022.818392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Fruit ripening is a highly complicated process, which is modulated by phytohormones, signal regulators and environmental factors playing in an intricate network that regulates ripening-related genes expression. Although transcriptomics is an effective tool to predict protein levels, protein abundances are also extensively affected by post-transcriptional and post-translational regulations. Here, we used RNA sequencing (RNA-seq) and tandem mass tag (TMT)-based quantitative proteomics to study the comprehensive mRNA and protein expression changes during fruit development and ripening in watermelon, a non-climacteric fruit. A total of 6,226 proteins were quantified, and the large number of quantitative proteins is comparable to proteomic studies in model organisms such as Oryza sativa L. and Arabidopsis. Base on our proteome methodology, integrative analysis of the transcriptome and proteome showed that the mRNA and protein levels were poorly correlated, and the correlation coefficients decreased during fruit ripening. Proteomic results showed that proteins involved in alternative splicing and the ubiquitin proteasome pathway were dynamically expressed during ripening. Furthermore, the spliceosome and proteasome were significantly enriched by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, suggesting that post-transcriptional and post-translational mechanisms might play important roles in regulation of fruit ripening-associated genes expression, which might account for the poor correlation between mRNAs and proteins during fruit ripening. Our comprehensive transcriptomic and proteomic data offer a valuable resource for watermelon research, and provide new insights into the molecular mechanisms underlying the complex regulatory networks of fruit ripening.
Collapse
|
19
|
Yun B, Ryu S, Kang M, Lee J, Yoo J, Kim Y, Oh S. Probiotic Lacticaseibacillus rhamnosus GG Increased Longevity and Resistance Against Foodborne Pathogens in Caenorhabditis elegans by Regulating MicroRNA miR-34. Front Cell Infect Microbiol 2022; 11:819328. [PMID: 35127565 PMCID: PMC8807481 DOI: 10.3389/fcimb.2021.819328] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigated the relation of probiotic activity of Lacticaseibacillus rhamnosus strain GG (LGG) and expression of microRNA to immune response and longevity in Caenorhabditis elegans host model. First, we evaluated the survival rate of C. elegans due to LGG exposure and bacterial colonization in the intestine. Next, the expression of mRNA and miRNA was analyzed in C. elegans exposure to LGG for 24 h using microarray. After exposure to LGG to C. elegans, colonized LGG was observed in the intestines of C. elegans and induced to extend lifespan. Moreover, persistent LGG in the intestine significantly enhanced the resistance of C. elegans exposed to both pathogenic bacteria and prolonged the lifespan of C. elegans. Transcriptome analysis indicated that LGG affected the expression levels of genes related to the innate immune response and upregulated the abundance of genes in multiple pathways of C. elegans, including Wnt signaling, TGF-beta signaling and mitogen-activated protein kinase (MAPK) pathways. In addition, qRT-PCR analysis confirmed that the expression of antibacterial genes was increased by LGG. Moreover, as the expression of microRNA miR-34 and immune-related pathways increased by exposure to LGG, the lifespan of C. elegans increased. However, in the miR-34 mutant C. elegans, the lifespan by LGG did not increase, so it was determined that miR-34 indirectly affects immune-related pathways. There was no significant difference in the expression of PMK-1 for LGG exposure in miR-34 mutants, suggesting that miR-34 may regulate PMK-1. In conclusion, we suggest that exposure of LGG to C. elegans enhances lifespan and resistance to food-borne pathogen infection by stimulating miR-34 and indirectly promoting PMK-1 activity.
Collapse
Affiliation(s)
- Bohyun Yun
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Sangdon Ryu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Minkyoung Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Juyeon Lee
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Jiseon Yoo
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
- *Correspondence: Younghoon Kim, ; Sangnam Oh,
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
- *Correspondence: Younghoon Kim, ; Sangnam Oh,
| |
Collapse
|
20
|
Cooper JF, Guasp RJ, Arnold ML, Grant BD, Driscoll M. Stress increases in exopher-mediated neuronal extrusion require lipid biosynthesis, FGF, and EGF RAS/MAPK signaling. Proc Natl Acad Sci U S A 2021; 118:e2101410118. [PMID: 34475208 PMCID: PMC8433523 DOI: 10.1073/pnas.2101410118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 06/23/2021] [Indexed: 01/08/2023] Open
Abstract
In human neurodegenerative diseases, neurons can transfer toxic protein aggregates to surrounding cells, promoting pathology via poorly understood mechanisms. In Caenorhabditis elegans, proteostressed neurons can expel neurotoxic proteins in large, membrane-bound vesicles called exophers. We investigated how specific stresses impact neuronal trash expulsion to show that neuronal exopher production can be markedly elevated by oxidative and osmotic stress. Unexpectedly, we also found that fasting dramatically increases exophergenesis. Mechanistic dissection focused on identifying nonautonomous factors that sense and activate the fasting-induced exopher response revealed that DAF16/FOXO-dependent and -independent processes are engaged. Fasting-induced exopher elevation requires the intestinal peptide transporter PEPT-1, lipid synthesis transcription factors Mediator complex MDT-15 and SBP-1/SREPB1, and fatty acid synthase FASN-1, implicating remotely initiated lipid signaling in neuronal trash elimination. A conserved fibroblast growth factor (FGF)/RAS/MAPK signaling pathway that acts downstream of, or in parallel to, lipid signaling also promotes fasting-induced neuronal exopher elevation. A germline-based epidermal growth factor (EGF) signal that acts through neurons is also required for exopher production. Our data define a nonautonomous network that links food availability changes to remote, and extreme, neuronal homeostasis responses relevant to aggregate transfer biology.
Collapse
Affiliation(s)
- Jason F Cooper
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Ryan J Guasp
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
- Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08901
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854;
| |
Collapse
|
21
|
Krassovsky K, Ghosh RP, Meyer BJ. Genome-wide profiling reveals functional interplay of DNA sequence composition, transcriptional activity, and nucleosome positioning in driving DNA supercoiling and helix destabilization in C. elegans. Genome Res 2021; 31:1187-1202. [PMID: 34168009 PMCID: PMC8256864 DOI: 10.1101/gr.270082.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
DNA topology and alternative DNA structures are implicated in regulating diverse biological processes. Although biomechanical properties of these structures have been studied extensively in vitro, characterization in vivo, particularly in multicellular organisms, is limited. We devised new methods to map DNA supercoiling and single-stranded DNA in Caenorhabditis elegans embryos and diapause larvae. To map supercoiling, we quantified the incorporation of biotinylated psoralen into DNA using high-throughput sequencing. To map single-stranded DNA, we combined permanganate treatment with genome-wide sequencing of induced double-stranded breaks. We found high levels of negative supercoiling at transcription start sites (TSSs) in embryos. GC-rich regions flanked by a sharp GC-to-AT transition delineate boundaries of supercoil propagation. In contrast to TSSs in embryos, TSSs in diapause larvae showed dramatic reductions in negative supercoiling without concomitant attenuation of transcription, suggesting developmental-stage-specific regulation. To assess whether alternative DNA structures control chromosome architecture and gene expression, we examined DNA supercoiling in the context of X-Chromosome dosage compensation. We showed that the condensin dosage compensation complex creates negative supercoils locally at its highest-occupancy binding sites but found no evidence for large-scale supercoiling domains along X Chromosomes. In contrast to transcription-coupled negative supercoiling, single-strandedness, which is most pronounced at transcript end sites, is dependent on high AT content and symmetrically positioned nucleosomes. We propose that sharp transitions in sequence composition at functional genomic elements constitute a common regulatory code and that DNA structure and propagation of torsional stress at regulatory elements are critical parameters in shaping important developmental events.
Collapse
Affiliation(s)
- Kristina Krassovsky
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3204, USA
| | - Rajarshi P Ghosh
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3204, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California 94720-3204, USA
| | - Barbara J Meyer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3204, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California 94720-3204, USA
| |
Collapse
|
22
|
Casasa S, Biddle JF, Koutsovoulos GD, Ragsdale EJ. Polyphenism of a Novel Trait Integrated Rapidly Evolving Genes into Ancestrally Plastic Networks. Mol Biol Evol 2021; 38:331-343. [PMID: 32931588 PMCID: PMC7826178 DOI: 10.1093/molbev/msaa235] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Developmental polyphenism, the ability to switch between phenotypes in response to environmental variation, involves the alternating activation of environmentally sensitive genes. Consequently, to understand how a polyphenic response evolves requires a comparative analysis of the components that make up environmentally sensitive networks. Here, we inferred coexpression networks for a morphological polyphenism, the feeding-structure dimorphism of the nematode Pristionchus pacificus. In this species, individuals produce alternative forms of a novel trait—moveable teeth, which in one morph enable predatory feeding—in response to environmental cues. To identify the origins of polyphenism network components, we independently inferred coexpression modules for more conserved transcriptional responses, including in an ancestrally nonpolyphenic nematode species. Further, through genome-wide analyses of these components across the nematode family (Diplogastridae) in which the polyphenism arose, we reconstructed how network components have changed. To achieve this, we assembled and resolved the phylogenetic context for five genomes of species representing the breadth of Diplogastridae and a hypothesized outgroup. We found that gene networks instructing alternative forms arose from ancestral plastic responses to environment, specifically starvation-induced metabolism and the formation of a conserved diapause (dauer) stage. Moreover, loci from rapidly evolving gene families were integrated into these networks with higher connectivity than throughout the rest of the P. pacificus transcriptome. In summary, we show that the modular regulatory outputs of a polyphenic response evolved through the integration of conserved plastic responses into networks with genes of high evolutionary turnover.
Collapse
Affiliation(s)
- Sofia Casasa
- Department of Biology, Indiana University, Bloomington, Bloomington, IN
| | - Joseph F Biddle
- Department of Biology, Indiana University, Bloomington, Bloomington, IN
| | | | - Erik J Ragsdale
- Department of Biology, Indiana University, Bloomington, Bloomington, IN
| |
Collapse
|
23
|
Sterken MG, van Wijk MH, Quamme EC, Riksen JAG, Carnell L, Mathies LD, Davies AG, Kammenga JE, Bettinger JC. Transcriptional analysis of the response of C. elegans to ethanol exposure. Sci Rep 2021; 11:10993. [PMID: 34040055 PMCID: PMC8155136 DOI: 10.1038/s41598-021-90282-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/07/2021] [Indexed: 11/30/2022] Open
Abstract
Ethanol-induced transcriptional changes underlie important physiological responses to ethanol that are likely to contribute to the addictive properties of the drug. We examined the transcriptional responses of Caenorhabditis elegans across a timecourse of ethanol exposure, between 30 min and 8 h, to determine what genes and genetic pathways are regulated in response to ethanol in this model. We found that short exposures to ethanol (up to 2 h) induced expression of metabolic enzymes involved in metabolizing ethanol and retinol, while longer exposure (8 h) had much more profound effects on the transcriptome. Several genes that are known to be involved in the physiological response to ethanol, including direct ethanol targets, were regulated at 8 h of exposure. This longer exposure to ethanol also resulted in the regulation of genes involved in cilia function, which is consistent with an important role for the effects of ethanol on cilia in the deleterious effects of chronic ethanol consumption in humans. Finally, we found that food deprivation for an 8-h period induced gene expression changes that were somewhat ameliorated by the presence of ethanol, supporting previous observations that worms can use ethanol as a calorie source.
Collapse
Affiliation(s)
- Mark G Sterken
- Laboratory of Nematology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Marijke H van Wijk
- Laboratory of Nematology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Elizabeth C Quamme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Box 980613, Richmond, VA, 23298, USA
| | - Joost A G Riksen
- Laboratory of Nematology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Lucinda Carnell
- Department of Biological Sciences, Central Washington University, Ellensburg, WA, 98926, USA
| | - Laura D Mathies
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Box 980613, Richmond, VA, 23298, USA
- Virginia Commonwealth University Alcohol Research Center, Richmond, VA, USA
| | - Andrew G Davies
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Box 980613, Richmond, VA, 23298, USA
- Virginia Commonwealth University Alcohol Research Center, Richmond, VA, USA
| | - Jan E Kammenga
- Laboratory of Nematology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Jill C Bettinger
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Box 980613, Richmond, VA, 23298, USA.
- Virginia Commonwealth University Alcohol Research Center, Richmond, VA, USA.
| |
Collapse
|
24
|
Dall KB, Havelund JF, Harvald EB, Witting M, Færgeman NJ. HLH-30-dependent rewiring of metabolism during starvation in C. elegans. Aging Cell 2021; 20:e13342. [PMID: 33724708 PMCID: PMC8045935 DOI: 10.1111/acel.13342] [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: 01/06/2020] [Revised: 02/08/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022] Open
Abstract
One of the most fundamental challenges for all living organisms is to sense and respond to alternating nutritional conditions in order to adapt their metabolism and physiology to promote survival and achieve balanced growth. Here, we applied metabolomics and lipidomics to examine temporal regulation of metabolism during starvation in wild‐type Caenorhabditis elegans and in animals lacking the transcription factor HLH‐30. Our findings show for the first time that starvation alters the abundance of hundreds of metabolites and lipid species in a temporal‐ and HLH‐30‐dependent manner. We demonstrate that premature death of hlh‐30 animals under starvation can be prevented by supplementation of exogenous fatty acids, and that HLH‐30 is required for complete oxidation of long‐chain fatty acids. We further show that RNAi‐mediated knockdown of the gene encoding carnitine palmitoyl transferase I (cpt‐1) only impairs survival of wild‐type animals and not of hlh‐30 animals. Strikingly, we also find that compromised generation of peroxisomes by prx‐5 knockdown renders hlh‐30 animals hypersensitive to starvation, which cannot be rescued by supplementation of exogenous fatty acids. Collectively, our observations show that mitochondrial functions are compromised in hlh‐30 animals and that hlh‐30 animals rewire their metabolism to largely depend on functional peroxisomes during starvation, underlining the importance of metabolic plasticity to maintain survival.
Collapse
Affiliation(s)
- Kathrine B. Dall
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences University of Southern Denmark Odense M Denmark
| | - Jesper F. Havelund
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences University of Southern Denmark Odense M Denmark
| | - Eva B. Harvald
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences University of Southern Denmark Odense M Denmark
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry Helmholtz Zentrum München Neuherberg Germany
- Metabolomics and Proteomics Core Helmholtz Zentrum München Neuherberg Germany
- Chair of Analytical Food Chemistry Technische Universität München Freising Germany
| | - Nils J. Færgeman
- Department of Biochemistry and Molecular Biology Villum Center for Bioanalytical Sciences University of Southern Denmark Odense M Denmark
| |
Collapse
|
25
|
Jiang Y, Liu Y, Zhang J. Mechanisms for the stimulatory effects of a five-component mixture of antibiotics in Microcystis aeruginosa at transcriptomic and proteomic levels. JOURNAL OF HAZARDOUS MATERIALS 2021; 406:124722. [PMID: 33296757 DOI: 10.1016/j.jhazmat.2020.124722] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Antibiotic contaminants could promote the formation of harmful cyanobacterial blooms through hormetic stimulation, but the mechanisms underlying these stimulatory effects remain unclear. This study investigated the biochemical, transcriptomic, and proteomic responses of a dominant bloom-forming cyanobacterium, Microcystis aeruginosa, to a five-component mixture of frequently detected antibiotics at current contamination levels. The growth rate of M. aeruginosa presented a U-shaped dose-response to 50-500 ng L-1 of mixed antibiotics. Alterations in the transcriptome of M. aeruginosa suggested the excitation of both photosynthesis and carbon metabolism, increasing energy generation in response to oxidative stress induced by low-dose antibiotics, and thus contributing to the significant (p < 0.05) increase in growth rate, Fv/Fm, and cell density. Comparison between transcriptomic and proteomic responses further confirmed the action mode of the mixed antibiotics. Proteins and their corresponding genes related to ROS scavenging, photosynthesis, carbon fixation, electron transport, oxidative phosphorylation, and biosynthesis, showed consistent expression tendencies in response to 200 ng L-1 of mixed antibiotics, which were credible action targets of mixed antibiotics in M. aeruginosa. Mixed antibiotics stimulated microcystin synthesis by upregulating a microcystin synthetase and its encoding gene (mcyC), which could increase the hazard of M. aeruginosa in aquatic environments.
Collapse
Affiliation(s)
- Yunhan Jiang
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, PR China
| | - Ying Liu
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, PR China.
| | - Jian Zhang
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, PR China
| |
Collapse
|
26
|
Fu T, Knittelfelder O, Geffard O, Clément Y, Testet E, Elie N, Touboul D, Abbaci K, Shevchenko A, Lemoine J, Chaumot A, Salvador A, Degli-Esposti D, Ayciriex S. Shotgun lipidomics and mass spectrometry imaging unveil diversity and dynamics in Gammarus fossarum lipid composition. iScience 2021; 24:102115. [PMID: 33615205 PMCID: PMC7881238 DOI: 10.1016/j.isci.2021.102115] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 01/14/2023] Open
Abstract
Sentinel species are playing an indispensable role in monitoring environmental pollution in aquatic ecosystems. Many pollutants found in water prove to be endocrine disrupting chemicals that could cause disruptions in lipid homeostasis in aquatic species. A comprehensive profiling of the lipidome of these species is thus an essential step toward understanding the mechanism of toxicity induced by pollutants. Both the composition and spatial distribution of lipids in freshwater crustacean Gammarus fossarum were extensively examined herein. The baseline lipidome of gammarids of different sex and reproductive stages was established by high throughput shotgun lipidomics. Spatial lipid mapping by high resolution mass spectrometry imaging led to the discovery of sulfate-based lipids in hepatopancreas and their accumulation in mature oocytes. A diverse and dynamic lipid composition in G. fossarum was uncovered, which deepens our understanding of the biochemical changes during development and which could serve as a reference for future ecotoxicological studies. Baseline lipidome profiling of G. fossarum of different sex and reproductive stages Spatial localization of lipids in gammarid tissue by mass spectrometry imaging SIMS imaging guided discovery of sulfate-based lipids in hepatopancreas epithelium Disclosure of a dynamic lipid composition in maturing female oocytes
Collapse
Affiliation(s)
- Tingting Fu
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Oskar Knittelfelder
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Olivier Geffard
- INRAE, UR RiverLy, Ecotoxicology Team, F-69625 Villeurbanne, France
| | - Yohann Clément
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Eric Testet
- Laboratoire de Biogenèse Membranaire (LBM), CNRS, University of Bordeaux, UMR 5200, F-33882 Villenave d'Ornon, France
| | - Nicolas Elie
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198, Gif-sur-Yvette, France
| | - David Touboul
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198, Gif-sur-Yvette, France
| | - Khedidja Abbaci
- INRAE, UR RiverLy, Ecotoxicology Team, F-69625 Villeurbanne, France
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Jerome Lemoine
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Arnaud Chaumot
- INRAE, UR RiverLy, Ecotoxicology Team, F-69625 Villeurbanne, France
| | - Arnaud Salvador
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | | | - Sophie Ayciriex
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
- Corresponding author
| |
Collapse
|
27
|
La Spina M, Contreras PS, Rissone A, Meena NK, Jeong E, Martina JA. MiT/TFE Family of Transcription Factors: An Evolutionary Perspective. Front Cell Dev Biol 2021; 8:609683. [PMID: 33490073 PMCID: PMC7815692 DOI: 10.3389/fcell.2020.609683] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Response and adaptation to stress are critical for the survival of all living organisms. The regulation of the transcriptional machinery is an important aspect of these complex processes. The members of the microphthalmia (MiT/TFE) family of transcription factors, apart from their involvement in melanocyte biology, are emerging as key players in a wide range of cellular functions in response to a plethora of internal and external stresses. The MiT/TFE proteins are structurally related and conserved through evolution. Their tissue expression and activities are highly regulated by alternative splicing, promoter usage, and posttranslational modifications. Here, we summarize the functions of MiT/TFE proteins as master transcriptional regulators across evolution and discuss the contribution of animal models to our understanding of the various roles of these transcription factors. We also highlight the importance of deciphering transcriptional regulatory mechanisms in the quest for potential therapeutic targets for human diseases, such as lysosomal storage disorders, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Martina La Spina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Pablo S Contreras
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alberto Rissone
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Naresh K Meena
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Eutteum Jeong
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Baugh LR, Hu PJ. Starvation Responses Throughout the Caenorhabditiselegans Life Cycle. Genetics 2020; 216:837-878. [PMID: 33268389 PMCID: PMC7768255 DOI: 10.1534/genetics.120.303565] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Caenorhabditis elegans survives on ephemeral food sources in the wild, and the species has a variety of adaptive responses to starvation. These features of its life history make the worm a powerful model for studying developmental, behavioral, and metabolic starvation responses. Starvation resistance is fundamental to life in the wild, and it is relevant to aging and common diseases such as cancer and diabetes. Worms respond to acute starvation at different times in the life cycle by arresting development and altering gene expression and metabolism. They also anticipate starvation during early larval development, engaging an alternative developmental program resulting in dauer diapause. By arresting development, these responses postpone growth and reproduction until feeding resumes. A common set of signaling pathways mediates systemic regulation of development in each context but with important distinctions. Several aspects of behavior, including feeding, foraging, taxis, egg laying, sleep, and associative learning, are also affected by starvation. A variety of conserved signaling, gene regulatory, and metabolic mechanisms support adaptation to starvation. Early life starvation can have persistent effects on adults and their descendants. With its short generation time, C. elegans is an ideal model for studying maternal provisioning, transgenerational epigenetic inheritance, and developmental origins of adult health and disease in humans. This review provides a comprehensive overview of starvation responses throughout the C. elegans life cycle.
Collapse
Affiliation(s)
- L Ryan Baugh
- Department of Biology, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708 and
| | - Patrick J Hu
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
29
|
Li HM, Liu P, Zhang XJ, Li LM, Jiang HY, Yan H, Hou FH, Chen JP. Combined proteomics and transcriptomics reveal the genetic basis underlying the differentiation of skin appendages and immunity in pangolin. Sci Rep 2020; 10:14566. [PMID: 32884035 PMCID: PMC7471334 DOI: 10.1038/s41598-020-71513-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/17/2020] [Indexed: 11/18/2022] Open
Abstract
Pangolin (Mains javanica) is an interesting endangered mammal with special morphological characteristics. Here, we applied proteomics and transcriptomics to explore the differentiation of pangolin skin appendages at two developmental stages and to compare gene expression profiles between abdomen hair and dorsal scale tissues. We identified 4,311 genes and 91 proteins differentially expressed between scale-type and hair-type tissue, of which 6 genes were shared by the transcriptome and proteome. Differentiation altered the abundance of hundreds of proteins and mRNA in the two types of skin appendages, many of which are involved in keratinocyte differentiation, epidermal cell differentiation, and multicellular organism development based on GO enrichment analysis, and FoxO, MAPK, and p53 signalling pathways based on KEGG enrichment analysis. DEGs in scale-type tissues were also significantly enriched in immune-related terms and pathways compared with that in hair-type tissues. Thus, we propose that pangolins have a normal skin innate immune system. Compared with the abdomen, the back skin of pangolins had more genes involved in the regulation of immune function, which may be an adaptive adjustment for the vulnerability of scaly skin to infection and injury. This investigation provides a scientific basis for the study of development and immunity of pangolin skin, which may be helpful in the protection of wild pangolin in China.
Collapse
Affiliation(s)
- Hui-Ming Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China
| | - Ping Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China
| | - Xiu-Juan Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China
| | - Lin-Miao Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China
| | - Hai-Ying Jiang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China
| | - Hua Yan
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou, Guangdong Province, China
| | - Fang-Hui Hou
- Guangdong Provincial Wildlife Rescue Centre, Guangzhou, Guangdong Province, China
| | - Jin-Ping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Science, Guangzhou, 510260, China.
| |
Collapse
|
30
|
Macedo F, Romanatto T, Gomes de Assis C, Buis A, Kowaltowski AJ, Aguilaniu H, Marques da Cunha F. Lifespan-extending interventions enhance lipid-supported mitochondrial respiration in Caenorhabditis elegans. FASEB J 2020; 34:9972-9981. [PMID: 32609395 DOI: 10.1096/fj.201901880r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/02/2020] [Accepted: 05/05/2020] [Indexed: 11/11/2022]
Abstract
Dietary restriction and reduced reproduction have been linked to long lifespans in the vast majority of species tested. Although decreased mitochondrial mass and/or function are hallmarks of aging, little is known about the mechanisms by which these organelles contribute to physiological aging or to the effects of lifespan-extending interventions, particularly with respect to oxidative phosphorylation and energy production. Here, we employed the nematode Caenorhabditis elegans to examine the effects of inhibition of germline proliferation and dietary restriction, both of which extend the lifespan of C. elegans, on mitochondrial respiratory activity in whole animals and isolated organelles. We found that oxygen consumption rates and mitochondrial mass were reduced in wild-type (WT) C. elegans subjected to bacterial deprivation (BD) compared with animals fed ad libitum (AL). In contrast, BD decreased the rate of oxygen uptake but not mitochondrial mass in germline-less glp-1(e2144ts) mutants. Interestingly, mitochondria isolated from animals subjected to BD and/or inhibition of germline proliferation showed no differences in complex I-mediated respiratory activity compared to control mitochondria, whereas both interventions enhanced the efficiency with which mitochondria utilized lipids as respiratory substrates. Notably, the combination of BD and inhibition of germline proliferation further increased mitochondrial lipid oxidation compared to either intervention alone. We also detected a striking correlation between lifespan extension in response to BD and/or inhibition of germline proliferation and the capacity of C. elegans to generate ATP from lipids. Our results thus suggest that the ability to oxidize lipids may be determinant in enhanced longevity.
Collapse
Affiliation(s)
- Felipe Macedo
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Talita Romanatto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Carolina Gomes de Assis
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alexia Buis
- Institut de Génomique Fonctionnelle de Lyon, Lyon, France
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Hugo Aguilaniu
- Institut de Génomique Fonctionnelle de Lyon, Lyon, France.,Instituto Serrapilheira, Rio de Janeiro, Brazil.,Centre National de la Recherche Scientifique, France
| | - Fernanda Marques da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Ji YJ, Ugolino J, Zhang T, Lu J, Kim D, Wang J. C9orf72/ALFA-1 controls TFEB/HLH-30-dependent metabolism through dynamic regulation of Rag GTPases. PLoS Genet 2020; 16:e1008738. [PMID: 32282804 PMCID: PMC7188304 DOI: 10.1371/journal.pgen.1008738] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/28/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrient utilization and energy metabolism are critical for the maintenance of cellular homeostasis. A mutation in the C9orf72 gene has been linked to the most common forms of neurodegenerative diseases that include amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we have identified an evolutionarily conserved function of C9orf72 in the regulation of the transcription factor EB (TFEB), a master regulator of autophagic and lysosomal genes that is negatively modulated by mTORC1. Loss of the C. elegans orthologue of C9orf72, ALFA-1, causes the nuclear translocation of HLH-30/TFEB, leading to activation of lipolysis and premature lethality during starvation-induced developmental arrest in C. elegans. A similar conserved pathway exists in human cells, in which C9orf72 regulates mTOR and TFEB signaling. C9orf72 interacts with and dynamically regulates the level of Rag GTPases, which are responsible for the recruitment of mTOR and TFEB on the lysosome upon amino acid signals. These results have revealed previously unknown functions of C9orf72 in nutrient sensing and metabolic pathways and suggest that dysregulation of C9orf72 functions could compromise cellular fitness under conditions of nutrient stress. An expansion of repeated nucleotides in the non-coding region of the C9orf72 gene has been linked to the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The repeat expansion leads to a reduced expression of the C9orf72 gene and loss of function of the C9orf72 protein may contribute to the pathogenesis. In this study, we identified a new mechanism through which C9orf72 influences nutrient sensing, autophagy, and metabolism. In the multi-cellular organism Caenorhabditis elegans, the C9orf72 orthologue regulates the activity of TFEB, a crucial transcriptional regulator of autophagic and lysosomal genes, through which the lipid metabolism and survival are influenced especially under nutrient stress conditions. The regulatory effect of C9orf72 on TFEB is conserved in mammals, and this is mediated by the dynamic regulation of the Rag GTPases by C9orf72. Given the critical role of the Rag GTPases in nutrient sensing and autophagy, we propose that the C9orf72 function is important for metabolic homeostasis in the cell and its deficiency can lead to compromised fitness under stress conditions.
Collapse
Affiliation(s)
- Yon Ju Ji
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Janet Ugolino
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Tao Zhang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Jiayin Lu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Dohoon Kim
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Jiou Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
32
|
Cruz-Corchado J, Ooi FK, Das S, Prahlad V. Global Transcriptome Changes That Accompany Alterations in Serotonin Levels in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2020; 10:1225-1246. [PMID: 31996358 PMCID: PMC7144078 DOI: 10.1534/g3.120.401088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/25/2020] [Indexed: 11/18/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT), is a phylogenetically ancient molecule best characterized as a neurotransmitter that modulates multiple aspects of mood and social cognition. The roles that 5-HT plays in normal and abnormal behavior are not fully understood but have been posited to be due to its common function as a 'defense signal'. However, 5-HT levels also systemically impact cell physiology, modulating cell division, migration, apoptosis, mitochondrial biogenesis, cellular metabolism and differentiation. Whether these diverse cellular effects of 5-HT also share a common basis is unclear. C. elegans provides an ideal system to interrogate the systemic effects of 5-HT, since lacking a blood-brain barrier, 5-HT synthesized and released by neurons permeates the organism to modulate neuronal as well as non-neuronal cells throughout the body. Here we used RNA-Seq to characterize the systemic changes in gene expression that occur in C. elegans upon altering 5-HT levels, and compared the transcriptomes to published datasets. We find that an acute increase in 5-HT is accompanied by a global decrease in gene expression levels, upregulation of genes involved in stress pathways, changes that significantly correlate with the published transcriptomes of animals that have activated defense and immune responses, and an increase in levels of phosphorylated eukaryotic initiation factor, eIF2α. In 5-HT deficient animals lacking tryptophan hydroxylase (tph-1(mg280)II) there is a net increase in gene expression, with an overrepresentation of genes related to development and chromatin. Surprisingly, the transcriptomes of animals with acute increases in 5-HT levels, and 5-HT deficiency do not overlap with transcriptomes of mutants with whom they share striking physiological resemblance. These studies are the first to catalog systemic transcriptome changes that occur upon alterations in 5-HT levels. They further show that in C. elegans changes in gene expression upon altering 5-HT levels, and changes in physiology, are not directly correlated.
Collapse
Affiliation(s)
- Johnny Cruz-Corchado
- Department of Biology, Aging Mind and Brain Initiative, Iowa Neuroscience Institute, 143 Biology Building, Iowa City, IA 52242-1324
| | - Felicia K Ooi
- Department of Biology, Aging Mind and Brain Initiative, Iowa Neuroscience Institute, 143 Biology Building, Iowa City, IA 52242-1324
| | - Srijit Das
- Department of Biology, Aging Mind and Brain Initiative, Iowa Neuroscience Institute, 143 Biology Building, Iowa City, IA 52242-1324
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative, Iowa Neuroscience Institute, 143 Biology Building, Iowa City, IA 52242-1324
| |
Collapse
|
33
|
Tao J, Hao Y, Li X, Yin H, Nie X, Zhang J, Xu B, Chen Q, Li B. Systematic Identification of Housekeeping Genes Possibly Used as References in Caenorhabditis elegans by Large-Scale Data Integration. Cells 2020; 9:E786. [PMID: 32213971 PMCID: PMC7140892 DOI: 10.3390/cells9030786] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
For accurate gene expression quantification, normalization of gene expression data against reliable reference genes is required. It is known that the expression levels of commonly used reference genes vary considerably under different experimental conditions, and therefore, their use for data normalization is limited. In this study, an unbiased identification of reference genes in Caenorhabditis elegans was performed based on 145 microarray datasets (2296 gene array samples) covering different developmental stages, different tissues, drug treatments, lifestyle, and various stresses. As a result, thirteen housekeeping genes (rps-23, rps-26, rps-27, rps-16, rps-2, rps-4, rps-17, rpl-24.1, rpl-27, rpl-33, rpl-36, rpl-35, and rpl-15) with enhanced stability were comprehensively identified by using six popular normalization algorithms and RankAggreg method. Functional enrichment analysis revealed that these genes were significantly overrepresented in GO terms or KEGG pathways related to ribosomes. Validation analysis using recently published datasets revealed that the expressions of newly identified candidate reference genes were more stable than the commonly used reference genes. Based on the results, we recommended using rpl-33 and rps-26 as the optimal reference genes for microarray and rps-2 and rps-4 for RNA-sequencing data validation. More importantly, the most stable rps-23 should be a promising reference gene for both data types. This study, for the first time, successfully displays a large-scale microarray data driven genome-wide identification of stable reference genes for normalizing gene expression data and provides a potential guideline on the selection of universal internal reference genes in C. elegans, for quantitative gene expression analysis.
Collapse
Affiliation(s)
- Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Xudong Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Xiner Nie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Jie Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Boying Xu
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Qiao Chen
- Scientific Research Office, Chongqing Normal University, Chongqing 401331, China;
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| |
Collapse
|
34
|
Haeussler S, Köhler F, Witting M, Premm MF, Rolland SG, Fischer C, Chauve L, Casanueva O, Conradt B. Autophagy compensates for defects in mitochondrial dynamics. PLoS Genet 2020; 16:e1008638. [PMID: 32191694 PMCID: PMC7135339 DOI: 10.1371/journal.pgen.1008638] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 04/06/2020] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
Compromising mitochondrial fusion or fission disrupts cellular homeostasis; however, the underlying mechanism(s) are not fully understood. The loss of C. elegans fzo-1MFN results in mitochondrial fragmentation, decreased mitochondrial membrane potential and the induction of the mitochondrial unfolded protein response (UPRmt). We performed a genome-wide RNAi screen for genes that when knocked-down suppress fzo-1MFN(lf)-induced UPRmt. Of the 299 genes identified, 143 encode negative regulators of autophagy, many of which have previously not been implicated in this cellular quality control mechanism. We present evidence that increased autophagic flux suppresses fzo-1MFN(lf)-induced UPRmt by increasing mitochondrial membrane potential rather than restoring mitochondrial morphology. Furthermore, we demonstrate that increased autophagic flux also suppresses UPRmt induction in response to a block in mitochondrial fission, but not in response to the loss of spg-7AFG3L2, which encodes a mitochondrial metalloprotease. Finally, we found that blocking mitochondrial fusion or fission leads to increased levels of certain types of triacylglycerols and that this is at least partially reverted by the induction of autophagy. We propose that the breakdown of these triacylglycerols through autophagy leads to elevated metabolic activity, thereby increasing mitochondrial membrane potential and restoring mitochondrial and cellular homeostasis.
Collapse
Affiliation(s)
- Simon Haeussler
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fabian Köhler
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Analytical Food Chemistry, Technische Universität München, Freising, Germany
| | - Madeleine F. Premm
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Christian Fischer
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
- Center for Integrated Protein Science, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Laetitia Chauve
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Olivia Casanueva
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Barbara Conradt
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
- Center for Integrated Protein Science, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London, United Kingdom
| |
Collapse
|
35
|
Yu S, Wang Z, Ding L, Yang L. The regulation of TFEB in lipid homeostasis of non-alcoholic fatty liver disease: Molecular mechanism and promising therapeutic targets. Life Sci 2020; 246:117418. [PMID: 32057899 DOI: 10.1016/j.lfs.2020.117418] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which is characterized by disruption of lipid homeostasis, has been the leading cause of chronic liver disease worldwide. However, currently there is no effective therapy for NAFLD. Consequently, it is extremely urgent to explore the specific and effective target functioned as lipids regulator during the pathological process of NAFLD for the drug development. Transcription factor EB (TFEB) plays a crucial role in the regulation of lipid homeostasis through linking autophagy to energy metabolism at the transcriptional level. In this review, we summarize the currently available information regarding the mediation of TFEB in lipid metabolism during the pathological process of NAFLD, and the specific regulatory mechanism of TFEB activity. We further recapitulate TFEB as a promising therapeutic target for NAFLD, primarily through the regulation of lipid homeostasis, energy metabolism as well as immune defense. A better understanding of these key issues will be helpful to promote the development of therapeutic agents which specifically target TFEB to halt or reverse the pathological progression of NAFLD.
Collapse
Affiliation(s)
- Shenglan Yu
- Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengtao Wang
- Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China.
| | - Li Yang
- Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
36
|
Helm ET, Curry SM, De Mille CM, Schweer WP, Burrough ER, Gabler NK. Impact of viral disease hypophagia on pig jejunal function and integrity. PLoS One 2020; 15:e0227265. [PMID: 31910236 PMCID: PMC6946155 DOI: 10.1371/journal.pone.0227265] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/14/2019] [Indexed: 01/19/2023] Open
Abstract
Pathogen challenges are often accompanied by reductions in feed intake, making it difficult to differentiate impacts of reduced feed intake from impacts of pathogen on various response parameters. Therefore, the objective of this study was to determine the impact of Porcine Reproductive and Respiratory Syndrome virus (PRRSV) and feed intake on parameters of jejunal function and integrity in growing pigs. Twenty-four pigs (11.34 ± 1.54 kg BW) were randomly selected and allotted to 1 of 3 treatments (n = 8 pigs/treatment): 1) PRRSV naïve, ad libitum fed (Ad), 2) PRRSV-inoculated, ad libitum fed (PRRS+), and 3) PRRSV naïve, pair-fed to the PRRS+ pigs' daily feed intake (PF). At 17 days post inoculation, all pigs were euthanized and the jejunum was collected for analysis. At days post inoculation 17, PRRS+ and PF pigs had decreased (P < 0.05) transepithelial resistance compared with Ad pigs; whereas fluorescein isothiocyanate-dextran 4 kDa permeability was not different among treatments. Active glucose transport was increased (P < 0.05) in PRRS+ and PF pigs compared with Ad pigs. Brush border carbohydrase activity was reduced in PRRS+ pigs compared with PF pigs for lactase (55%; P = 0.015), sucrase (37%; P = 0.002), and maltase (30%; P = 0.015). For all three carbohydrases, Ad pigs had activities intermediate that of PRRS+ and PF pigs. The mRNA abundance of the tight junction proteins claudin 2, claudin 3, claudin 4, occludin, and zonula occludens-1 were reduced in PRRS+ pigs compared with Ad pigs; however, neither the total protein abundance nor the cellular compartmentalization of these tight junction proteins differed among treatments. Taken together, this study demonstrates that the changes that occur to intestinal epithelium structure, function, and integrity during a systemic PRRSV challenge can be partially explained by reductions in feed intake. Further, long term adaptation to PRRSV challenge and caloric restriction does reduce intestinal transepithelial resistance but does not appear to reduce the integrity of tight junction protein complexes.
Collapse
Affiliation(s)
- Emma T. Helm
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Shelby M. Curry
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Carson M. De Mille
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Wesley P. Schweer
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Eric R. Burrough
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Nicholas K. Gabler
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| |
Collapse
|
37
|
Bouagnon AD, Lin L, Srivastava S, Liu CC, Panda O, Schroeder FC, Srinivasan S, Ashrafi K. Intestinal peroxisomal fatty acid β-oxidation regulates neural serotonin signaling through a feedback mechanism. PLoS Biol 2019; 17:e3000242. [PMID: 31805041 PMCID: PMC6917301 DOI: 10.1371/journal.pbio.3000242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 12/17/2019] [Accepted: 11/15/2019] [Indexed: 02/02/2023] Open
Abstract
The ability to coordinate behavioral responses with metabolic status is fundamental to the maintenance of energy homeostasis. In numerous species including Caenorhabditis elegans and mammals, neural serotonin signaling regulates a range of food-related behaviors. However, the mechanisms that integrate metabolic information with serotonergic circuits are poorly characterized. Here, we identify metabolic, molecular, and cellular components of a circuit that links peripheral metabolic state to serotonin-regulated behaviors in C. elegans. We find that blocking the entry of fatty acyl coenzyme As (CoAs) into peroxisomal β-oxidation in the intestine blunts the effects of neural serotonin signaling on feeding and egg-laying behaviors. Comparative genomics and metabolomics revealed that interfering with intestinal peroxisomal β-oxidation results in a modest global transcriptional change but significant changes to the metabolome, including a large number of changes in ascaroside and phospholipid species, some of which affect feeding behavior. We also identify body cavity neurons and an ether-a-go-go (EAG)-related potassium channel that functions in these neurons as key cellular components of the circuitry linking peripheral metabolic signals to regulation of neural serotonin signaling. These data raise the possibility that the effects of serotonin on satiety may have their origins in feedback, homeostatic metabolic responses from the periphery.
Collapse
Affiliation(s)
- Aude D. Bouagnon
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Lin Lin
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Shubhi Srivastava
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Chung-Chih Liu
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Oishika Panda
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Frank C. Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Supriya Srinivasan
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
38
|
Dall KB, Færgeman NJ. Metabolic regulation of lifespan from a C. elegans perspective. GENES & NUTRITION 2019; 14:25. [PMID: 31428207 PMCID: PMC6694653 DOI: 10.1186/s12263-019-0650-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Decline of cellular functions especially cognitive is a major deficit that arises with age in humans. Harnessing the strengths of small and genetic tractable model systems has revealed key conserved regulatory biochemical and signaling pathways that control aging. Here, we review some of the key signaling and biochemical pathways that coordinate aging processes with special emphasis on Caenorhabditis elegans as a model system and discuss how nutrients and metabolites can regulate lifespan by coordinating signaling and epigenetic programs. We focus on central nutrient-sensing pathways such as mTOR and insulin/insulin-like growth factor signaling and key transcription factors including the conserved basic helix-loop-helix transcription factor HLH-30/TFEB.
Collapse
Affiliation(s)
- Kathrine B. Dall
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Nils J. Færgeman
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
39
|
Abstract
Starvation is among the most ancient of selection pressures, driving evolution of a robust arsenal of starvation survival defenses. In order to survive starvation stress, organisms must be able to curtail anabolic processes during starvation and judiciously activate catabolic pathways. Although the activation of metabolic defenses in response to nutrient deprivation is an obvious component of starvation survival, less appreciated is the importance of the ability to recover from starvation upon re-exposure to nutrients. In order for organisms to successfully recover from starvation, cells must be kept in a state of ready so that upon the return of nutrients, activities such as growth and reproduction can be resumed. Critical to this state of ready is the lysosome, an organelle that provides essential signals of nutrient sufficiency to cell growth-activating pathways in the fed state. In this issue, Murphy and colleagues provide evidence that exposure of Caenorhabditis elegans roundworms to 2 simple nutrients, glucose and the polyunsaturated fatty acid linoleate, is able to render lysosomal function competent to activate key downstream starvation recovery pathways, bypassing the need for a master transcriptional regulator of lysosomes. These findings provide a quantum leap forward in our understanding of the cellular determinants that permit organisms to survive cycles of feast and famine. Organisms require elaborate systems to defend against nutrient stress. This Primer explores recent evidence that the transcription factor TFEB, a master regulator of starvation defences, also primes animals for recovery from starvation once food becomes available.
Collapse
Affiliation(s)
- Alexander A. Soukas
- Department of Medicine, Diabetes Unit, and Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- * E-mail:
| | - Ben Zhou
- Department of Medicine, Diabetes Unit, and Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| |
Collapse
|
40
|
Murphy JT, Liu H, Ma X, Shaver A, Egan BM, Oh C, Boyko A, Mazer T, Ang S, Khopkar R, Javaheri A, Kumar S, Jiang X, Ory D, Mani K, Matkovich SJ, Kornfeld K, Diwan A. Simple nutrients bypass the requirement for HLH-30 in coupling lysosomal nutrient sensing to survival. PLoS Biol 2019; 17:e3000245. [PMID: 31086360 PMCID: PMC6516633 DOI: 10.1371/journal.pbio.3000245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
Lysosomes are ubiquitous acidified organelles that degrade intracellular and extracellular material trafficked via multiple pathways. Lysosomes also sense cellular nutrient levels to regulate target of rapamycin (TOR) kinase, a signaling enzyme that drives growth and suppresses activity of the MiT/TFE family of transcription factors that control biogenesis of lysosomes. In this study, we subjected worms lacking basic helix–loop–helix transcription factor 30 (hlh-30), the Caenorhabditis elegans MiT/TFE ortholog, to starvation followed by refeeding to understand how this pathway regulates survival with variable nutrient supply. Loss of HLH-30 markedly impaired survival in starved larval worms and recovery upon refeeding bacteria. Remarkably, provision of simple nutrients in a completely defined medium (C. elegans maintenance medium [CeMM]), specifically glucose and linoleic acid, restored lysosomal acidification, TOR activation, and survival with refeeding despite the absence of HLH-30. Worms deficient in lysosomal lipase 2 (lipl-2), a lysosomal enzyme that is transcriptionally up-regulated in starvation in an HLH-30–dependent manner, also demonstrated increased mortality with starvation–refeeding that was partially rescued with glucose, suggesting a critical role for LIPL-2 in lipid metabolism under starvation. CeMM induced transcription of vacuolar proton pump subunits in hlh-30 mutant worms, and knockdown of vacuolar H+-ATPase 12 (vha-12) and its upstream regulator, nuclear hormone receptor 31 (nhr-31), abolished the rescue with CeMM. Loss of Ras-related GTP binding protein C homolog 1 RAGC-1, the ortholog for mammalian RagC/D GTPases, conferred starvation–refeeding lethality, and RAGC-1 overexpression was sufficient to rescue starved hlh-30 mutant worms, demonstrating a critical need for TOR activation with refeeding. These results show that HLH-30 activation is critical for sustaining survival during starvation–refeeding stress via regulating TOR. Glucose and linoleic acid bypass the requirement for HLH-30 in coupling lysosome nutrient sensing to survival. Lysosomes play a central role in coupling the nutrient state of the cell to growth and survival decisions. This study uncovers a critical role for HLH-30, the nematode ortholog of the mammalian MiT/TFE family of master regulators of lysosome biogenesis, in survival under starvation and refeeding conditions. Refeeding simple nutrients bypasses the requirement for HLH-30 to permit survival.
Collapse
Affiliation(s)
- John T. Murphy
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Haiyan Liu
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- John Cochran VA Medical Center, St. Louis, Missouri, United States of America
| | - Xiucui Ma
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- John Cochran VA Medical Center, St. Louis, Missouri, United States of America
| | - Alex Shaver
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Brian M. Egan
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Clara Oh
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexander Boyko
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Travis Mazer
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Samuel Ang
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rohan Khopkar
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ali Javaheri
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sandeep Kumar
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xuntian Jiang
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Daniel Ory
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kartik Mani
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- John Cochran VA Medical Center, St. Louis, Missouri, United States of America
| | - Scot J. Matkovich
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Abhinav Diwan
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- John Cochran VA Medical Center, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
41
|
Denzel MS, Lapierre LR, Mack HID. Emerging topics in C. elegans aging research: Transcriptional regulation, stress response and epigenetics. Mech Ageing Dev 2018; 177:4-21. [PMID: 30134144 PMCID: PMC6696993 DOI: 10.1016/j.mad.2018.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/13/2022]
Abstract
Key discoveries in aging research have been made possible with the use of model organisms. Caenorhabditis elegans is a short-lived nematode that has become a well-established system to study aging. The practicality and powerful genetic manipulations associated with this metazoan have revolutionized our ability to understand how organisms age. 25 years after the publication of the discovery of the daf-2 gene as a genetic modifier of lifespan, C. elegans remains as relevant as ever in the quest to understand the process of aging. Nematode aging research has proven useful in identifying transcriptional regulators, small molecule signals, cellular mechanisms, epigenetic modifications associated with stress resistance and longevity, and lifespan-extending compounds. Here, we review recent discoveries and selected topics that have emerged in aging research using this incredible little worm.
Collapse
Affiliation(s)
- Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| | | |
Collapse
|