1
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in Caenorhabditis elegans. eLife 2024; 13:RP97503. [PMID: 39636673 PMCID: PMC11620748 DOI: 10.7554/elife.97503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here, we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in Caenorhabditis elegans in the intestine. We identify an FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calcium-dependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
| | - Drew Young
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
2
|
Turmel-Couture S, Martel PO, Beaulieu L, Lechasseur X, Fotso Dzuna LV, Narbonne P. Bidirectional transfer of a small membrane-impermeable molecule between the Caenorhabditis elegans intestine and germline. J Biol Chem 2024; 300:107963. [PMID: 39510179 DOI: 10.1016/j.jbc.2024.107963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) is a positive regulator of cell proliferation often upregulated in cancer. Its Caenorhabditis elegans ortholog MPK-1 stimulates germline stem cell (GSC) proliferation nonautonomously from the intestine or somatic gonad. How MPK-1 can perform this task from either of these two tissues however remains unclear. We reasoned that somatic MPK-1 activity could lead to the generation of proproliferative small molecules that could transfer from the intestine and/or somatic gonad to the germline. Here, in support of this hypothesis, we demonstrate that a significant fraction of the small membrane-impermeable fluorescent molecule, 5-carboxyfluorescein, transfers to the germline after its microinjection in the animal's intestine. The larger part of this transfer targets oocytes and requires the germline receptor mediated endocytosis 2 (RME-2) yolk receptor. A minor quantity of the dye is however distributed independently from RME-2 and more widely in the animal, including the distal germline, gonadal sheath, coelomocytes, and hypodermis. We further show that the intestine-to-germline transfer efficiency of this RME-2 independent fraction does not vary together with GSC proliferation rates or MPK-1 activity. Therefore, if germline proliferation was influenced by small membrane-impermeable molecules generated in the intestine, it is unlikely that proliferation would be regulated at the level of molecule transfer rate. Finally, we show that conversely, a similar fraction of germline injected 5-carboxyfluorescein transfers to the intestine, demonstrating transfer bidirectionality. Altogether, our results establish the possibility of an intestine-to-germline signaling axis mediated by small membrane-impermeable molecules that could promote GSC proliferation cell nonautonomously downstream of MPK-1 activity.
Collapse
Affiliation(s)
- Sarah Turmel-Couture
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Pier-Olivier Martel
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Lucie Beaulieu
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Xavier Lechasseur
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | | | - Patrick Narbonne
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada.
| |
Collapse
|
3
|
Driesschaert B, Mergan L, Lucci C, Simon C, Santos D, De Groef L, Temmerman L. The role of phagocytic cells in aging: insights from vertebrate and invertebrate models. Biogerontology 2024; 25:1301-1314. [PMID: 39168928 DOI: 10.1007/s10522-024-10131-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
While the main role of phagocytic scavenger cells consists of the neutralization and elimination of pathogens, they also keep the body fluids clean by taking up and breaking down waste material. Since a build-up of waste is thought to contribute to the aging process, these cells become particularly pertinent in the research field of aging. Nevertheless, a direct link between their scavenging functions and the aging process has yet to be established. Integrative approaches involving various model organisms hold promise to elucidate this potential, but are lagging behind since the diversity and evolutionary relationship of these cells across animal species remain unclear. In this perspective, we review the current knowledge associating phagocytic scavenger cells with aging in vertebrate and invertebrate animals, as well as put forward important questions for further exploration. Additionally, we highlight future challenges and propose a constructive approach for tackling them.
Collapse
Affiliation(s)
- Brecht Driesschaert
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Lucas Mergan
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Cristiano Lucci
- Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, Naamsestraat 61 - Box 2464, B-3000, Leuven, Belgium
| | - Caroline Simon
- Molecular Developmental Physiology and Signal Transduction, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Dulce Santos
- Molecular Developmental Physiology and Signal Transduction, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, Naamsestraat 61 - Box 2464, B-3000, Leuven, Belgium
| | - Liesbet Temmerman
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium.
| |
Collapse
|
4
|
Taylor SKB, Hartman JH, Gupta BP. The neurotrophic factor MANF regulates autophagy and lysosome function to promote proteostasis in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2024; 121:e2403906121. [PMID: 39418305 PMCID: PMC11513987 DOI: 10.1073/pnas.2403906121] [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: 02/24/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
The conserved mesencephalic astrocyte-derived neurotrophic factor (MANF) is known for protecting dopaminergic neurons and functioning in various other tissues. Previously, we showed that Caenorhabditis elegans manf-1 null mutants exhibit defects such as increased endoplasmic reticulum (ER) stress, dopaminergic neurodegeneration, and abnormal protein aggregation. These findings suggest an essential role for MANF in cellular processes. However, the mechanisms by which intracellular and extracellular MANF regulate broader cellular functions remain unclear. We report a unique mechanism of action for MANF-1 that involves the transcription factor HLH-30/TFEB-mediated signaling to regulate autophagy and lysosomal function. Multiple transgenic strains overexpressing MANF-1 showed extended lifespan of animals, reduced protein aggregation, and improved neuronal survival. Using fluorescently tagged MANF-1, we observed tissue-specific localization of the protein, which was dependent on the ER retention signal. Further subcellular analysis showed that MANF-1 localizes within cells to the lysosomes and utilizes the endosomal pathway. Consistent with the lysosomal localization, our transcriptomic study of MANF-1 and analyses of autophagy regulators demonstrated that MANF-1 promotes proteostasis by regulating autophagic flux and lysosomal activity. Collectively, our findings establish MANF as a critical regulator of stress response, proteostasis, and aging.
Collapse
Affiliation(s)
| | - Jessica H. Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC29425
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC29425
| | - Bhagwati P. Gupta
- Department of Biology, McMaster University, Hamilton, ONL8S 4K1, Canada
| |
Collapse
|
5
|
Lo JY, Adam KM, Garrison JL. Neuropeptide inactivation regulates egg-laying behavior to influence reproductive health in Caenorhabditis elegans. Curr Biol 2024; 34:4715-4728.e4. [PMID: 39395417 DOI: 10.1016/j.cub.2024.09.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/11/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024]
Abstract
Neural communication requires both fast-acting neurotransmitters and neuromodulators that function on slower timescales to communicate. Endogenous bioactive peptides, often called "neuropeptides," comprise the largest and most diverse class of neuromodulators that mediate crosstalk between the brain and peripheral tissues to regulate physiology and behaviors conserved across the animal kingdom. Neuropeptide signaling can be terminated through receptor binding and internalization or degradation by extracellular enzymes called neuropeptidases. Inactivation by neuropeptidases can shape the dynamics of signaling in vivo by specifying both the duration of signaling and the anatomic path neuropeptides can travel before they are degraded. For most neuropeptides, the identity of the relevant inactivating peptidase(s) is unknown. Here, we established a screening platform in C. elegans utilizing mass spectrometry-based peptidomics to discover neuropeptidases and simultaneously profile the in vivo specificity of these enzymes against each of more than 250 endogenous peptides. We identified NEP-2, a worm ortholog of the mammalian peptidase neprilysin-2, and demonstrated that it regulates specific neuropeptides, including those in the egg-laying circuit. We found that NEP-2 is required in muscle cells to regulate signals from neurons to modulate both behavior and health in the reproductive system. Taken together, our results demonstrate that peptidases, which are an important node of regulation in neuropeptide signaling, affect the dynamics of signaling to impact behavior, physiology, and aging.
Collapse
Affiliation(s)
- Jacqueline Y Lo
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Katelyn M Adam
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA
| | - Jennifer L Garrison
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA; Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16th Street, San Francisco, CA 94158, USA; Center for Healthy Aging in Women, 8001 Redwood Boulevard, Novato, CA 94945, USA; Productive Health Global Consortium, 8001 Redwood Boulevard, Novato, CA 94945, USA.
| |
Collapse
|
6
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587937. [PMID: 39345448 PMCID: PMC11429608 DOI: 10.1101/2024.04.03.587937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in C. elegans in the intestine. We identify a FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calciumdependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
| | - Drew Young
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
7
|
Emser J, Wernet N, Hetzer B, Wohlmann E, Fischer R. The cysteine-rich virulence factor NipA of Arthrobotrys flagrans interferes with cuticle integrity of Caenorhabditis elegans. Nat Commun 2024; 15:5795. [PMID: 38987250 PMCID: PMC11237121 DOI: 10.1038/s41467-024-50096-4] [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: 10/13/2023] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Animals protect themself from microbial attacks by robust skins or a cuticle as in Caenorhabditis elegans. Nematode-trapping fungi, like Arthrobotrys flagrans, overcome the cuticle barrier and colonize the nematode body. While lytic enzymes are important for infection, small-secreted proteins (SSPs) without enzymatic activity, emerge as crucial virulence factors. Here, we characterized NipA (nematode induced protein) which A. flagrans secretes at the penetration site. In the absence of NipA, A. flagrans required more time to penetrate C. elegans. Heterologous expression of the fungal protein in the epidermis of C. elegans led to blister formation. NipA contains 13 cysteines, 12 of which are likely to form disulfide bridges, and the remaining cysteine was crucial for blister formation. We hypothesize that NipA interferes with cuticle integrity to facilitate fungal entry. Genome-wide expression analyses of C. elegans expressing NipA revealed mis-regulation of genes associated with extracellular matrix (ECM) maintenance and innate immunity.
Collapse
Affiliation(s)
- Jennifer Emser
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Nicole Wernet
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Birgit Hetzer
- Max Rubner-Institut (MRI) - Federal Research Institute of Nutrition and Food, Haid-und-Neu-Strasse 9, Karlsruhe, 76131, Germany
| | - Elke Wohlmann
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Reinhard Fischer
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany.
| |
Collapse
|
8
|
Ohse VA, Klotz LO, Priebs J. Copper Homeostasis in the Model Organism C. elegans. Cells 2024; 13:727. [PMID: 38727263 PMCID: PMC11083455 DOI: 10.3390/cells13090727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Cellular and organismic copper (Cu) homeostasis is regulated by Cu transporters and Cu chaperones to ensure the controlled uptake, distribution and export of Cu ions. Many of these processes have been extensively investigated in mammalian cell culture, as well as in humans and in mammalian model organisms. Most of the human genes encoding proteins involved in Cu homeostasis have orthologs in the model organism, Caenorhabditis elegans (C. elegans). Starting with a compilation of human Cu proteins and their orthologs, this review presents an overview of Cu homeostasis in C. elegans, comparing it to the human system, thereby establishing the basis for an assessment of the suitability of C. elegans as a model to answer mechanistic questions relating to human Cu homeostasis.
Collapse
Affiliation(s)
| | - Lars-Oliver Klotz
- Nutrigenomics Section, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, 07743 Jena, Germany;
| | - Josephine Priebs
- Nutrigenomics Section, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, 07743 Jena, Germany;
| |
Collapse
|
9
|
Zajac M, Mukherjee S, Anees P, Oettinger D, Henn K, Srikumar J, Zou J, Saminathan A, Krishnan Y. A mechanism of lysosomal calcium entry. SCIENCE ADVANCES 2024; 10:eadk2317. [PMID: 38354239 PMCID: PMC10866540 DOI: 10.1126/sciadv.adk2317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
Lysosomal calcium (Ca2+) release is critical to cell signaling and is mediated by well-known lysosomal Ca2+ channels. Yet, how lysosomes refill their Ca2+ remains hitherto undescribed. Here, from an RNA interference screen in Caenorhabditis elegans, we identify an evolutionarily conserved gene, lci-1, that facilitates lysosomal Ca2+ entry in C. elegans and mammalian cells. We found that its human homolog TMEM165, previously designated as a Ca2+/H+ exchanger, imports Ca2+ pH dependently into lysosomes. Using two-ion mapping and electrophysiology, we show that TMEM165, hereafter referred to as human LCI, acts as a proton-activated, lysosomal Ca2+ importer. Defects in lysosomal Ca2+ channels cause several neurodegenerative diseases, and knowledge of lysosomal Ca2+ importers may provide previously unidentified avenues to explore the physiology of Ca2+ channels.
Collapse
Affiliation(s)
- Matthew Zajac
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Sourajit Mukherjee
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Palapuravan Anees
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Daphne Oettinger
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Katharine Henn
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Jainaha Srikumar
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Junyi Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Anand Saminathan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
10
|
Fanelli MJ, Welsh CM, Lui DS, Smulan LJ, Walker AK. Immunity-linked genes are stimulated by a membrane stress pathway linked to Golgi function and the ARF-1 GTPase. SCIENCE ADVANCES 2023; 9:eadi5545. [PMID: 38055815 PMCID: PMC10699786 DOI: 10.1126/sciadv.adi5545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Infection response and other immunity-linked genes (ILGs) were first named in Caenorhabditis elegans-based expression after pathogen challenge, but many are also up-regulated when lipid metabolism is perturbed. Why pathogen attack and metabolic changes both increase ILGs is unclear. We find that ILGs are activated when phosphatidylcholine (PC) levels change in membranes of secretory organelles in C. elegans. RNAi targeting of the ADP-ribosylation factor arf-1, which disrupts the Golgi and secretory function, also activates ILGs. Low PC limits ARF-1 function, suggesting a mechanism for ILG activation via lipid metabolism, as part of a membrane stress response acting outside the ER. RNAi of selected ILGs uncovered defects in the secretion of two GFP reporters and the accumulation of a pathogen-responsive complement C1r/C1s, Uegf, Bmp1 (CUB) domain fusion protein. Our data argue that up-regulation of some ILGs is a coordinated response to changes in trafficking and may act to counteract stress on secretory function.
Collapse
Affiliation(s)
- Matthew J. Fanelli
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester, MA, USA
| | - Christofer M. Welsh
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester, MA, USA
- Morningside School of Biomedical Sciences, UMASS Chan Medical School, Worcester, MA, USA
| | - Dominique S. Lui
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester, MA, USA
| | - Lorissa J. Smulan
- Department of Medicine, UMASS Chan Medical School, Worcester, MA, USA
| | - Amy K. Walker
- Program in Molecular Medicine, UMASS Chan Medical School, Worcester, MA, USA
| |
Collapse
|
11
|
Gengyo-Ando K, Tateyama M, Mitani S, Ando H, Nakai J. A humanized Caenorhabditis elegans model for studying pathogenic mutations in VPS45, a protein essential for membrane trafficking, associated with severe congenital neutropenia. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.001052. [PMID: 38089934 PMCID: PMC10714219 DOI: 10.17912/micropub.biology.001052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 02/01/2024]
Abstract
VPS45, one of the essential membrane trafficking factors, has been identified as a cause of severe congenital neutropenia 5 (SCN5), but its pathophysiological role remains unknown. Here, we developed a humanized C. elegans model for three pathogenic VPS45 variants. We found that wild-type human VPS45 functionally complemented the loss of C. elegans VPS-45 , and the pathogenic human VPS45 variants functioned almost normally with respect to larval development and endocytosis in C. elegans . These results suggest that SCN5-associated mutations have little effect on the core function of VPS45, and/or that the degree of VPS45 requirement varies, depending on the cell/tissue.
Collapse
Affiliation(s)
- Keiko Gengyo-Ando
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Minoru Tateyama
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Shohei Mitani
- Physiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideki Ando
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Junichi Nakai
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| |
Collapse
|
12
|
Koop K, Yuan W, Tessadori F, Rodriguez-Polanco WR, Grubbs J, Zhang B, Osmond M, Graham G, Sawyer S, Conboy E, Vetrini F, Treat K, Płoski R, Pienkowski VM, Kłosowska A, Fieg E, Krier J, Mallebranche C, Alban Z, Aldinger KA, Ritter D, Macnamara E, Sullivan B, Herriges J, Alaimo JT, Helbig C, Ellis CA, van Eyk C, Gecz J, Farrugia D, Osei-Owusu I, Adès L, van den Boogaard MJ, Fuchs S, Bakker J, Duran K, Dawson ZD, Lindsey A, Huang H, Baldridge D, Silverman GA, Grant BD, Raizen D, van Haaften G, Pak SC, Rehmann H, Schedl T, van Hasselt P. Macrocephaly and developmental delay caused by missense variants in RAB5C. Hum Mol Genet 2023; 32:3063-3077. [PMID: 37552066 PMCID: PMC10586195 DOI: 10.1093/hmg/ddad130] [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: 03/30/2023] [Revised: 07/06/2023] [Accepted: 07/29/2023] [Indexed: 08/09/2023] Open
Abstract
Rab GTPases are important regulators of intracellular vesicular trafficking. RAB5C is a member of the Rab GTPase family that plays an important role in the endocytic pathway, membrane protein recycling and signaling. Here we report on 12 individuals with nine different heterozygous de novo variants in RAB5C. All but one patient with missense variants (n = 9) exhibited macrocephaly, combined with mild-to-moderate developmental delay. Patients with loss of function variants (n = 2) had an apparently more severe clinical phenotype with refractory epilepsy and intellectual disability but a normal head circumference. Four missense variants were investigated experimentally. In vitro biochemical studies revealed that all four variants were damaging, resulting in increased nucleotide exchange rate, attenuated responsivity to guanine exchange factors and heterogeneous effects on interactions with effector proteins. Studies in C. elegans confirmed that all four variants were damaging in vivo and showed defects in endocytic pathway function. The variant heterozygotes displayed phenotypes that were not observed in null heterozygotes, with two shown to be through a dominant negative mechanism. Expression of the human RAB5C variants in zebrafish embryos resulted in defective development, further underscoring the damaging effects of the RAB5C variants. Our combined bioinformatic, in vitro and in vivo experimental studies and clinical data support the association of RAB5C missense variants with a neurodevelopmental disorder characterized by macrocephaly and mild-to-moderate developmental delay through disruption of the endocytic pathway.
Collapse
Affiliation(s)
- Klaas Koop
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, 3584 EA, The Netherlands
| | - Weimin Yuan
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Federico Tessadori
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Wilmer R Rodriguez-Polanco
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jeremy Grubbs
- Department of Neurology and the Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bo Zhang
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Matt Osmond
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| | - Gail Graham
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| | - Sarah Sawyer
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| | - Erin Conboy
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Francesco Vetrini
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kayla Treat
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Rafal Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, 02-106, Poland
| | - Victor Murcia Pienkowski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, 02-106, Poland
- Marseille Medical Genetics U1251, Aix Marseille University, Marseille, 13005, France
| | - Anna Kłosowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Elizabeth Fieg
- Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Joel Krier
- Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Coralie Mallebranche
- Unité d'Onco-Hémato-Immunologie pédiatrique, CHU d’Angers, Angers, 49933, France
| | - Ziegler Alban
- Service de génétique, CHU d’Angers, Angers, 49933, France
| | - Kimberly A Aldinger
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Deborah Ritter
- Department of Pediatrics, Oncology Section, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ellen Macnamara
- Undiagnosed Diseases Program Translational Laboratory, NHGRI, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bonnie Sullivan
- Division of Clinical Genetics, Department of Pediatrics, Children's Mercy-Kansas City, Kansas City, MO, 64108, USA
| | - John Herriges
- Department of Pathology and Laboratory Medicine, Children's Mercy-Kansas City, Kansas City, MO, 64108, USA
| | - Joseph T Alaimo
- Department of Pathology and Laboratory Medicine, Children's Mercy-Kansas City, Kansas City, MO, 64108, USA
| | - Catherine Helbig
- The Epilepsy Neurogenetics Initiative, Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Colin A Ellis
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia PA, 19104, USA
| | - Clare van Eyk
- Robinson Research Institute, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5006, Australia
| | - Jozef Gecz
- Robinson Research Institute, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5006, Australia
| | | | - Ikeoluwa Osei-Owusu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Lesley Adès
- Department of Clinical Genetics, The Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, 2145, Australia
| | - Marie-Jose van den Boogaard
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3584EA, The Netherlands
| | - Sabine Fuchs
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, 3584 EA, The Netherlands
| | - Jeroen Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Karen Duran
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Zachary D Dawson
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Anika Lindsey
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Huiyan Huang
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Dustin Baldridge
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Gary A Silverman
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - David Raizen
- Department of Neurology and the Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gijs van Haaften
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3584EA, The Netherlands
| | - Stephen C Pak
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Holger Rehmann
- Department of Energy and Biotechnology, Flensburg University of Applied Sciences, 24943, Flensburg, Germany
| | - Tim Schedl
- Departments of Pediatrics and Genetics, C. elegans Model Organism Screening Center, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Peter van Hasselt
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, 3584 EA, The Netherlands
| |
Collapse
|
13
|
Park J, Xie Y, Miller KG, De Camilli P, Yogev S. End-binding protein 1 promotes specific motor-cargo association in the cell body prior to axonal delivery of dense core vesicles. Curr Biol 2023; 33:3851-3864.e7. [PMID: 37586371 PMCID: PMC10529979 DOI: 10.1016/j.cub.2023.07.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/25/2023] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Axonal transport is key to neuronal function. Efficient transport requires specific motor-cargo association in the soma, yet the mechanisms regulating this early step remain poorly understood. We found that EBP-1, the C. elegans ortholog of the canonical-microtubule-end-binding protein EB1, promotes the specific association between kinesin-3/KIF1A/UNC-104 and dense core vesicles (DCVs) prior to their axonal delivery. Using single-neuron, in vivo labeling of endogenous cargo and EBs, we observed reduced axonal abundance and reduced secretion of DCV cargo, but not other KIF1A/UNC-104 cargoes, in ebp-1 mutants. This reduction could be traced back to fewer exit events from the cell body, where EBP-1 colocalized with the DCV sorting machinery at the trans Golgi, suggesting that this is the site of EBP-1 function. EBP-1 calponin homology (CH) domain was required for directing microtubule growth on the Golgi, and mammalian EB1 interacted with KIF1A in an EBH-domain-dependent manner. Loss- and gain-of-function experiments suggest a model in which both kinesin-3 binding and guidance of microtubule growth at the trans Golgi by EBP-1 promote motor-cargo association at sites of DCV biogenesis. In support of this model, tethering either EBP-1 or a kinesin-3/KIF1A/UNC-104-interacting domain from an unrelated protein to the Golgi restored the axonal abundance of DCV proteins in ebp-1 mutants. These results uncover an unexpected role for a microtubule-associated protein and provide insights into how specific kinesin-3 cargo is delivered to the axon.
Collapse
Affiliation(s)
- Junhyun Park
- Department of Neuroscience, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA
| | - Yi Xie
- Department of Neuroscience, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA
| | - Kenneth G Miller
- Genetic Models of Disease Laboratory, Oklahoma Medical Research Foundation, 825 N. E. 13th St, Oklahoma City, OK 73104, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA
| | - Shaul Yogev
- Department of Neuroscience, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
14
|
Zhang J, Jiang Z, Chen C, Yao L, Gao Z, Cheng Z, Yan Y, Liu H, Shi A. Age-associated decline in RAB-10 efficacy impairs intestinal barrier integrity. NATURE AGING 2023; 3:1107-1127. [PMID: 37640905 DOI: 10.1038/s43587-023-00475-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 07/24/2023] [Indexed: 08/31/2023]
Abstract
The age-related decline in the ability of the intestinal barrier to maintain selective permeability can lead to various physiological disturbances. Adherens junctions play a vital role in regulating intestinal permeability, and their proper assembly is contingent upon endocytic recycling. However, how aging affects the recycling efficiency and, consequently, the integrity of adherens junctions remains unclear. Here we show that RAB-10/Rab10 functionality is reduced during senescence, leading to impaired adherens junctions in the Caenorhabditis elegans intestine. Mechanistic analysis reveals that SDPN-1/PACSINs is upregulated in aging animals, suppressing RAB-10 activation by competing with DENN-4/GEF. Consistently, SDPN-1 knockdown alleviates age-related abnormalities in adherens junction integrity and intestinal barrier permeability. Of note, the inhibitory effect of SDPN-1 on RAB-10 requires KGB-1/JUN kinase, which presumably enhances the potency of SDPN-1 by altering its oligomerization state. Together, by examining age-associated changes in endocytic recycling, our study sheds light on how aging can impact intestinal barrier permeability.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Zongyan Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Changling Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Longfeng Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Zihang Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yanling Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Anbing Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Liu J, Zhang P, Zheng Z, Afridi MI, Zhang S, Wan Z, Zhang X, Stingelin L, Wang Y, Tu H. GABAergic signaling between enteric neurons and intestinal smooth muscle promotes innate immunity and gut defense in Caenorhabditis elegans. Immunity 2023; 56:1515-1532.e9. [PMID: 37437538 DOI: 10.1016/j.immuni.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/06/2023] [Accepted: 06/07/2023] [Indexed: 07/14/2023]
Abstract
The nervous system is critical for intestinal homeostasis and function, but questions remain regarding its impact on gut immune defense. By screening the major neurotransmitters of C. elegans, we found that γ-aminobutyric acid (GABA) deficiency enhanced susceptibility to pathogenic Pseudomonas aeruginosa PA14 infection. GABAergic signaling between enteric neurons and intestinal smooth muscle promoted gut defense in a PMK-1/p38-dependent, but IIS/DAF-16- and DBL-1/TGF-β-independent, pathway. Transcriptomic profiling revealed that the neuropeptide, FLP-6, acted downstream of enteric GABAergic signaling. Further data determined that FLP-6 was expressed and secreted by intestinal smooth muscle cells and functioned as a paracrine molecule on the intestinal epithelium. FLP-6 suppressed the transcription factors ZIP-10 and KLF-1 that worked in parallel and converged to the PMK-1/p38 pathway in the intestinal epithelia for innate immunity and gut defense. Collectively, these findings uncover an enteric neuron-muscle-epithelium axis that may be evolutionarily conserved in higher organisms.
Collapse
Affiliation(s)
- Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Pei Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Zhongfan Zheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Muhammad Irfan Afridi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Shan Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Zhiqing Wan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Xiumei Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Lukas Stingelin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Yirong Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, Hunan, China.
| |
Collapse
|
16
|
Peng JY, Liu X, Zeng XT, Hao Y, Zhang JH, Li Q, Tong XJ. Early pheromone perception remodels neurodevelopment and accelerates neurodegeneration in adult C. elegans. Cell Rep 2023; 42:112598. [PMID: 37289584 DOI: 10.1016/j.celrep.2023.112598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
Age-associated neurodegenerative disorders such as Parkinson's and Alzheimer's diseases are mainly caused by protein aggregation. The etiologies of these neurodegenerative diseases share a chemical environment. However, how chemical cues modulate neurodegeneration remains unclear. Here, we found that in Caenorhabditis elegans, exposure to pheromones in the L1 stage accelerates neurodegeneration in adults. Perception of pheromones ascr#3 and ascr#10 is mediated by chemosensory neurons ASK and ASI. ascr#3 perceived by G protein-coupled receptor (GPCR) DAF-38 in ASK activates glutamatergic transmission into AIA interneurons. ascr#10 perceived by GPCR STR-2 in ASI activates the secretion of neuropeptide NLP-1, which binds to the NPR-11 receptor in AIA. Activation of both ASI and ASK is required and sufficient to remodel neurodevelopment via AIA, which triggers insulin-like signaling and inhibits autophagy in adult neurons non-cell-autonomously. Our work reveals how pheromone perception at the early developmental stage modulates neurodegeneration in adults and provides insights into how the external environment impacts neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing-Yi Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuqing Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xian-Ting Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yue Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia-Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Lingang Laboratory, Shanghai 200031, China
| | - Qian Li
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Department of Anatomy and Physiology, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health in Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia-Jing Tong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
17
|
Mergan L, Driesschaert B, Temmerman L. Endocytic coelomocytes are required for lifespan extension by axenic dietary restriction. PLoS One 2023; 18:e0287933. [PMID: 37368903 DOI: 10.1371/journal.pone.0287933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
A rather peculiar but very potent means of achieving longevity is through axenic dietary restriction (ADR), where animals feed on (semi-)defined culture medium in absence of any other lifeform. The little knowledge we already have on ADR is mainly derived from studies using the model organism Caenorhabditis elegans, where ADR more than doubles organismal lifespan. What is underlying this extreme longevity so far remains enigmatic, as ADR seems distinct from other forms of DR and bypasses well-known longevity factors. We here focus first on CUP-4, a protein present in the coelomocytes, which are endocytic cells with a presumed immune function. Our results show that loss of cup-4 or of the coelomocytes affects ADR-mediated longevity to a similar extent. As the coelomocytes have been suggested to have an immune function, we then investigated different central players of innate immune signalling, but could prove no causal links with axenic lifespan extension. We propose that future research focuses further on the role of the coelomocytes in endocytosis and recycling in the context of longevity.
Collapse
Affiliation(s)
- Lucas Mergan
- Department of Biology, Animal Physiology and Neurobiology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Brecht Driesschaert
- Department of Biology, Animal Physiology and Neurobiology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Liesbet Temmerman
- Department of Biology, Animal Physiology and Neurobiology, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
18
|
Dejima K, Imae R, Suehiro Y, Yoshida K, Mitani S. An endomembrane zinc transporter negatively regulates systemic RNAi in Caenorhabditis elegans. iScience 2023; 26:106930. [PMID: 37305693 PMCID: PMC10250833 DOI: 10.1016/j.isci.2023.106930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/18/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Double-stranded RNA (dsRNA) regulates gene expression in a sequence-dependent manner. In Caenorhabditis elegans, dsRNA spreads through the body and leads to systemic RNA silencing. Although several genes involved in systemic RNAi have been genetically identified, molecules that mediate systemic RNAi remain largely unknown. Here, we identified ZIPT-9, a C. elegans homolog of ZIP9/SLC39A9, as a broad-spectrum negative regulator of systemic RNAi. We showed that RSD-3, SID-3, and SID-5 genetically act in parallel for efficient RNAi, and that zipt-9 mutants suppress the RNAi defects of all the mutants. Analysis of a complete set of deletion mutants for SLC30 and SLC39 family genes revealed that only zipt-9 mutants showed altered RNAi activity. Based on these results and our analysis using transgenic Zn2+ reporters, we propose that ZIPT-9-dependent Zn2+ homeostasis, rather than overall cytosolic Zn2+, modulates systemic RNAi activity. Our findings reveal a previously unknown function of zinc transporters in negative RNAi regulation.
Collapse
Affiliation(s)
- Katsufumi Dejima
- Department of Physiology, Tokyo Women’s Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Rieko Imae
- Department of Physiology, Tokyo Women’s Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Yuji Suehiro
- Department of Physiology, Tokyo Women’s Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Keita Yoshida
- Department of Physiology, Tokyo Women’s Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Shohei Mitani
- Department of Physiology, Tokyo Women’s Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
19
|
Ono M, Matsushita K, Maega S, Asano N, Matsunaga Y, Bito T, Iwasaki T, Kawano T. The G protein-coupled receptor neuropeptide receptor-15 modulates larval development via the transforming growth factor-β DAF-7 protein in Caenorhabditis elegans. Biochem Biophys Res Commun 2023; 660:28-34. [PMID: 37060828 DOI: 10.1016/j.bbrc.2023.03.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
G protein-coupled receptors (GPCRs) are a major class of membrane receptors that modulate a wide range of physiological functions. These receptors transmit extracellular signals, including secreted bioactive peptides, to intracellular signaling pathways. The nematode Caenorhabditis elegans has FMRFamide-like peptides, which are one of the most diverse neuropeptide families, some of which modulate larval development through GPCRs. In this study, we identified the GPCR neuropeptide receptor (NPR)-15, which modulates C. elegans larval development. Our molecular genetic analyses indicated the following: 1) NPR-15 mainly functions in ASI neurons, which predominantly regulate larval development, 2) NPR-15 interacts with GPA-4, a C. elegans Gα subunit, and 3) NPR-15, along with GPA-4, modulates larval development by regulating the production and secretion of the transforming growth factor-β (TGF-β)-like protein DAF-7. The present study is the first report to demonstrate the importance of a GPCR to the direct regulation of a TGF-β-like protein.
Collapse
Affiliation(s)
- Masahiro Ono
- Department of Bioresources Science, The United Graduate School of Agriculture, Japan
| | - Kenjiro Matsushita
- Department of Agricultural Science, Graduate School of Sustainability Science, Japan
| | - Sho Maega
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Naoto Asano
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | | | - Tomohiro Bito
- Department of Bioresources Science, The United Graduate School of Agriculture, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Japan; Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Takashi Iwasaki
- Department of Bioresources Science, The United Graduate School of Agriculture, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Japan; Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Bioresources Science, The United Graduate School of Agriculture, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Japan; Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan.
| |
Collapse
|
20
|
Hao Y, Liu H, Zeng XT, Wang Y, Zeng WX, Qian KY, Li L, Chi MX, Gao S, Hu Z, Tong XJ. UNC-43/CaMKII-triggered anterograde signals recruit GABA ARs to mediate inhibitory synaptic transmission and plasticity at C. elegans NMJs. Nat Commun 2023; 14:1436. [PMID: 36918518 PMCID: PMC10015018 DOI: 10.1038/s41467-023-37137-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Disturbed inhibitory synaptic transmission has functional impacts on neurodevelopmental and psychiatric disorders. An essential mechanism for modulating inhibitory synaptic transmission is alteration of the postsynaptic abundance of GABAARs, which are stabilized by postsynaptic scaffold proteins and recruited by presynaptic signals. However, how GABAergic neurons trigger signals to transsynaptically recruit GABAARs remains elusive. Here, we show that UNC-43/CaMKII functions at GABAergic neurons to recruit GABAARs and modulate inhibitory synaptic transmission at C. elegans neuromuscular junctions. We demonstrate that UNC-43 promotes presynaptic MADD-4B/Punctin secretion and NRX-1α/Neurexin surface delivery. Together, MADD-4B and NRX-1α recruit postsynaptic NLG-1/Neuroligin and stabilize GABAARs. Further, the excitation of GABAergic neurons potentiates the recruitment of NLG-1-stabilized-GABAARs, which depends on UNC-43, MADD-4B, and NRX-1. These data all support that UNC-43 triggers MADD-4B and NRX-1α, which act as anterograde signals to recruit postsynaptic GABAARs. Thus, our findings elucidate a mechanism for pre- and postsynaptic communication and inhibitory synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Yue Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Xian-Ting Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ya Wang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wan-Xin Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Kang-Ying Qian
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ming-Xuan Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Shangbang Gao
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Xia-Jing Tong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
21
|
de Lima FMR, Abrahão I, Pentagna N, Carneiro K. Gradual specialization of phagocytic ameboid cells may have impaired regenerative capacities in metazoan lineages. Dev Dyn 2023; 252:343-362. [PMID: 36205096 DOI: 10.1002/dvdy.543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/08/2022] Open
Abstract
Animal regeneration is a fascinating field of research that has captured the attention of many generations of scientists. Among the cellular mechanisms underlying tissue and organ regeneration, we highlight the role of phagocytic ameboid cells (PACs). Beyond their ability to engulf nutritional particles, microbes, and apoptotic cells, their involvement in regeneration has been widely documented. It has been extensively described that, at least in part, animal regenerative mechanisms rely on PACs that serve as a hub for a range of critical physiological functions, both in health and disease. Considering the phylogenetics of PAC evolution, and the loss and gain of nutritional, immunological, and regenerative potential across Metazoa, we aim to discuss when and how phagocytic activity was first co-opted to regenerative tissue repair. We propose that the gradual specialization of PACs during metazoan derivation may have contributed to the loss of regenerative potential in animals, with critical impacts on potential translational strategies for regenerative medicine.
Collapse
Affiliation(s)
- Felipe Matheus Ribeiro de Lima
- Laboratory of Cellular Proliferation and Differentiation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Developmental Biology, Postgraduate Program in Morphological Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabella Abrahão
- Laboratory of Cellular Proliferation and Differentiation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathalia Pentagna
- Laboratory of Cellular Proliferation and Differentiation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate Program in Medicine (Pathological Anatomy), Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Katia Carneiro
- Laboratory of Cellular Proliferation and Differentiation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Developmental Biology, Postgraduate Program in Morphological Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate Program in Medicine (Pathological Anatomy), Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Une R, Kageyama N, Ono M, Matsunaga Y, Iwasaki T, Kawano T. The FMRFamide-like peptide FLP-1 modulates larval development by regulating the production and secretion of the insulin-like peptide DAF-28 in Caenorhabditis elegans. Biosci Biotechnol Biochem 2023; 87:171-178. [PMID: 36507740 DOI: 10.1093/bbb/zbac187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/12/2022] [Indexed: 12/14/2022]
Abstract
The FMRFamide-like peptides (FLPs) are conserved in both free-living and parasitic nematodes. This molecular genetic study verified the relevance of the flp-1 gene, which is conserved in many nematode species, to the larval development of the free-living soil nematode Caenorhabditis elegans. Using C. elegans as a model, we found that: (1) FLP-1 suppressed larval development, resulting in diapause; (2) the secretion of FLP-1, which is produced in AVK head neurons, was suppressed by the presence of food (Escherichia coli) as an environmental factor to continue larval development; (3) the FLP-1 reduced the production and secretion of DAF-28, which is produced in ASI head neurons and is the predominant insulin-like peptide (INS) present. FLP-1 is conserved in many species of plant-parasitic root-knot nematodes that cause severe damage to crops. Therefore, our findings may provide insight into the development of new nematicides that can disturb their infection and development.
Collapse
Affiliation(s)
- Risako Une
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori, Tottori 680-8553, Japan
| | - Natsumi Kageyama
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori, Tottori 680-8553, Japan
| | - Masahiro Ono
- Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | | | - Takashi Iwasaki
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori, Tottori 680-8553, Japan
- Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori, Tottori 680-8553, Japan
- Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
23
|
Davis K, Mitchell C, Weissenfels O, Bai J, Raizen DM, Ailion M, Topalidou I. G protein-coupled receptor kinase-2 (GRK-2) controls exploration through neuropeptide signaling in Caenorhabditis elegans. PLoS Genet 2023; 19:e1010613. [PMID: 36652499 PMCID: PMC9886303 DOI: 10.1371/journal.pgen.1010613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Animals alter their behavior in manners that depend on environmental conditions as well as their developmental and metabolic states. For example, C. elegans is quiescent during larval molts or during conditions of satiety. By contrast, worms enter an exploration state when removed from food. Sensory perception influences movement quiescence (defined as a lack of body movement), as well as the expression of additional locomotor states in C. elegans that are associated with increased or reduced locomotion activity, such as roaming (exploration behavior) and dwelling (local search). Here we find that movement quiescence is enhanced, and exploration behavior is reduced in G protein-coupled receptor kinase grk-2 mutant animals. grk-2 was previously shown to act in chemosensation, locomotion, and egg-laying behaviors. Using neuron-specific rescuing experiments, we show that GRK-2 acts in multiple ciliated chemosensory neurons to control exploration behavior. grk-2 acts in opposite ways from the cGMP-dependent protein kinase gene egl-4 to control movement quiescence and exploration behavior. Analysis of mutants with defects in ciliated sensory neurons indicates that grk-2 and the cilium-structure mutants act in the same pathway to control exploration behavior. We find that GRK-2 controls exploration behavior in an opposite manner from the neuropeptide receptor NPR-1 and the neuropeptides FLP-1 and FLP-18. Finally, we show that secretion of the FLP-1 neuropeptide is negatively regulated by GRK-2 and that overexpression of FLP-1 reduces exploration behavior. These results define neurons and molecular pathways that modulate movement quiescence and exploration behavior.
Collapse
Affiliation(s)
- Kristen Davis
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Center for Excellence in Environmental Toxicology (CEET), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Christo Mitchell
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Olivia Weissenfels
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Jihong Bai
- Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - David M. Raizen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael Ailion
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Irini Topalidou
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
24
|
The p38 MAPK/PMK-1 Pathway Is Required for Resistance to Nocardia farcinica Infection in Caenorhabditis elegance. Pathogens 2022; 11:pathogens11101071. [PMID: 36297128 PMCID: PMC9609018 DOI: 10.3390/pathogens11101071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/02/2022] Open
Abstract
Nocardia farcinica is an opportunistic pathogen that causes nocardiosis primarily in patients with compromised immune systems. In this study, we used the genetically tractable organism Caenorhabditis elegans as a model to study the innate immune responses to N. farcinica infection. We found that unlike other pathogenic bacteria such as Pseudomonas aeruginosa and Staphylococcus aureus, N. farcinica failed to kill adult worms. In another words, adult worms exposed to N. farcinica exhibited a normal lifespan, compared with those fed the standard laboratory food bacterium Escherichia coli OP50. Interestingly, deletion of three core genes (pmk-1, nsy-1 and sek-1) in the p38 MAPK/PMK-1 pathway reduced the survival of worm exposure to N. farcinica, highlighting a crucial role of this pathway for C. elegans in resistance to N. farcinica. Furthermore, our results revealed that N. farcinica exposure up-regulated the level of PMK-1 phosphorylation. The activation of PMK-1 promoted nuclear translocation of a transcription factor SKN-1/Nrf2, which in turn mediated N. farcinica infection resistance in C. elegans. Our results provide an excellent example that the integrity of immune system is key aspect for counteract with pathogenesis of N. farcinica.
Collapse
|
25
|
Kageyama N, Nose M, Ono M, Matsunaga Y, Iwasaki T, Kawano T. The FMRFamide-like peptide FLP-2 is involved in the modulation of larval development and adult lifespan by regulating the secretion of the insulin-like peptide INS-35 in Caenorhabditis elegans. Biosci Biotechnol Biochem 2022; 86:1231-1239. [PMID: 35786701 DOI: 10.1093/bbb/zbac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/05/2022] [Indexed: 11/12/2022]
Abstract
In the animal kingdom, neuropeptides regulate diverse physiological functions. In invertebrates, FMRFamide and its related peptides, a family of neuropeptides, play an important role as neurotransmitters. The FMRFamide-like peptides (FLPs) are one of the most diverse neuropeptide families and are conserved in nematodes. Our screen for flp genes of the free-living soil nematode Caenorhabditis elegans revealed that the flp-2 gene is involved in larval development. The gene is also conserved in plant-parasitic root-knot nematodes. Our molecular genetic analyses of the C. elegans flp-2 gene demonstrated as follows: 1) the production and secretion of FLP-2, produced in the head neurons, are controlled by environmental factors (growth density and food); 2) the FLP-2 is involved in not only larval development but also adult lifespan by regulating the secretion of one of the insulin-like peptides INS-35, produced in the intestine. These findings provide new insight into the development of new nematicides.
Collapse
Affiliation(s)
- Natsumi Kageyama
- Department of Agricultural Science, Graduate School of Sustainability Science
| | - Masayo Nose
- Department of Agricultural Science, Graduate School of Sustainability Science
| | - Masahiro Ono
- Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | | | - Takashi Iwasaki
- Department of Agricultural Science, Graduate School of Sustainability Science.,Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Agricultural Science, Graduate School of Sustainability Science.,Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
26
|
Savini M, Folick A, Lee YT, Jin F, Cuevas A, Tillman MC, Duffy JD, Zhao Q, Neve IA, Hu PW, Yu Y, Zhang Q, Ye Y, Mair WB, Wang J, Han L, Ortlund EA, Wang MC. Lysosome lipid signalling from the periphery to neurons regulates longevity. Nat Cell Biol 2022; 24:906-916. [PMID: 35681008 PMCID: PMC9203275 DOI: 10.1038/s41556-022-00926-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 04/25/2022] [Indexed: 12/13/2022]
Abstract
Lysosomes are key cellular organelles that metabolize extra- and intracellular substrates. Alterations in lysosomal metabolism are implicated in ageing-associated metabolic and neurodegenerative diseases. However, how lysosomal metabolism actively coordinates the metabolic and nervous systems to regulate ageing remains unclear. Here we report a fat-to-neuron lipid signalling pathway induced by lysosomal metabolism and its longevity-promoting role in Caenorhabditis elegans. We discovered that induced lysosomal lipolysis in peripheral fat storage tissue upregulates the neuropeptide signalling pathway in the nervous system to promote longevity. This cell-non-autonomous regulation is mediated by a specific polyunsaturated fatty acid, dihomo-γ-linolenic acid, and LBP-3 lipid chaperone protein transported from the fat storage tissue to neurons. LBP-3 binds to dihomo-γ-linolenic acid, and acts through NHR-49 nuclear receptor and NLP-11 neuropeptide in neurons to extend lifespan. These results reveal lysosomes as a signalling hub to coordinate metabolism and ageing, and lysosomal signalling mediated inter-tissue communication in promoting longevity.
Collapse
Affiliation(s)
- Marzia Savini
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Andrew Folick
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Yi-Tang Lee
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Integrative Program of Molecular and Biochemical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Feng Jin
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - André Cuevas
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew C Tillman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathon D Duffy
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Qian Zhao
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Isaiah A Neve
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Pei-Wen Hu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qinghao Zhang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - William B Mair
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.,Center of Epigenetics and Disease Prevention, Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA. .,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
27
|
Chai CM, Torkashvand M, Seyedolmohadesin M, Park H, Venkatachalam V, Sternberg PW. Interneuron control of C. elegans developmental decision-making. Curr Biol 2022; 32:2316-2324.e4. [PMID: 35447086 DOI: 10.1016/j.cub.2022.03.077] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/13/2022] [Accepted: 03/30/2022] [Indexed: 01/18/2023]
Abstract
Natural environments are highly dynamic, and this complexity challenges animals to accurately integrate external cues to shape their responses. Adaptive developmental plasticity enables organisms to remodel their physiology, morphology, and behavior to better suit the predicted future environment and ultimately enhance their ecological success.1 Understanding how an animal generates a neural representation of current and forecasted environmental conditions and converts these circuit computations into a predictive adaptive physiological response may provide fundamental insights into the molecular and cellular basis of decision-making over developmentally relevant timescales. Although it is known that sensory cues usually trigger the developmental switch and that downstream inter-tissue signaling pathways enact the alternative developmental phenotype, the integrative neural mechanisms that transduce external inputs into effector pathways are less clear.2,3 In adverse environments, Caenorhabditis elegans larvae can enter a stress-resistant diapause state with arrested metabolism and reproductive physiology.4 Amphid sensory neurons feed into both rapid chemotactic and short-term foraging mode decisions, mediated by amphid and pre-motor interneurons, as well as the long-term diapause entry decision. Here, we identify amphid interneurons that integrate pheromone cues and propagate this information via a neuropeptidergic pathway to influence larval developmental fate, bypassing the pre-motor system. AIA interneuron-derived FLP-2 neuropeptide signaling promotes reproductive growth, and AIA activity is suppressed by pheromones. FLP-2 signaling is inhibited by upstream glutamatergic transmission via the metabotropic receptor MGL-1 and mediated by the broadly expressed neuropeptide G-protein-coupled receptor NPR-30. Thus, metabotropic signaling allows the reuse of parts of a sensory system for a decision with a distinct timescale.
Collapse
Affiliation(s)
- Cynthia M Chai
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA.
| | - Mahdi Torkashvand
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| | | | - Heenam Park
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA
| | - Vivek Venkatachalam
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA.
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA.
| |
Collapse
|
28
|
Abouelezz A, Almeida-Souza L. The mammalian endocytic cytoskeleton. Eur J Cell Biol 2022; 101:151222. [DOI: 10.1016/j.ejcb.2022.151222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022] Open
|
29
|
Florman JT, Alkema MJ. Co-transmission of neuropeptides and monoamines choreograph the C. elegans escape response. PLoS Genet 2022; 18:e1010091. [PMID: 35239681 PMCID: PMC8932558 DOI: 10.1371/journal.pgen.1010091] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/18/2022] [Accepted: 02/11/2022] [Indexed: 11/19/2022] Open
Abstract
Co-localization and co-transmission of neurotransmitters and neuropeptides is a core property of neural signaling across species. While co-transmission can increase the flexibility of cellular communication, understanding the functional impact on neural dynamics and behavior remains a major challenge. Here we examine the role of neuropeptide/monoamine co-transmission in the orchestration of the C. elegans escape response. The tyraminergic RIM neurons, which coordinate distinct motor programs of the escape response, also co-express the neuropeptide encoding gene flp-18. We find that in response to a mechanical stimulus, flp-18 mutants have defects in locomotory arousal and head bending that facilitate the omega turn. We show that the induction of the escape response leads to the release of FLP-18 neuropeptides. FLP-18 modulates the escape response through the activation of the G-protein coupled receptor NPR-5. FLP-18 increases intracellular calcium levels in neck and body wall muscles to promote body bending. Our results show that FLP-18 and tyramine act in different tissues in both a complementary and antagonistic manner to control distinct motor programs during different phases of the C. elegans flight response. Our study reveals basic principles by which co-transmission of monoamines and neuropeptides orchestrate in arousal and behavior in response to stress.
Collapse
Affiliation(s)
- Jeremy T. Florman
- Department of Neurobiology, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Mark J. Alkema
- Department of Neurobiology, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
30
|
Jomaa A, Gamerdinger M, Hsieh HH, Wallisch A, Chandrasekaran V, Ulusoy Z, Scaiola A, Hegde RS, Shan SO, Ban N, Deuerling E. Mechanism of signal sequence handover from NAC to SRP on ribosomes during ER-protein targeting. Science 2022; 375:839-844. [PMID: 35201867 PMCID: PMC7612438 DOI: 10.1126/science.abl6459] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The nascent polypeptide-associated complex (NAC) interacts with newly synthesized proteins at the ribosomal tunnel exit and competes with the signal recognition particle (SRP) to prevent mistargeting of cytosolic and mitochondrial polypeptides to the endoplasmic reticulum (ER). How NAC antagonizes SRP and how this is overcome by ER targeting signals are unknown. Here, we found that NAC uses two domains with opposing effects to control SRP access. The core globular domain prevented SRP from binding to signal-less ribosomes, whereas a flexibly attached domain transiently captured SRP to permit scanning of nascent chains. The emergence of an ER-targeting signal destabilized NAC's globular domain and facilitated SRP access to the nascent chain. These findings elucidate how NAC hands over the signal sequence to SRP and imparts specificity of protein localization.
Collapse
Affiliation(s)
- Ahmad Jomaa
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, 8093 Zurich, Switzerland
| | - Martin Gamerdinger
- Department of Biology, Molecular Microbiology, University of Konstanz, 78457 Konstanz, Germany
| | - Hao-Hsuan Hsieh
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Annalena Wallisch
- Department of Biology, Molecular Microbiology, University of Konstanz, 78457 Konstanz, Germany
| | | | - Zeynel Ulusoy
- Department of Biology, Molecular Microbiology, University of Konstanz, 78457 Konstanz, Germany
| | - Alain Scaiola
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Shu-ou Shan
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Nenad Ban
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, 8093 Zurich, Switzerland
| | - Elke Deuerling
- Department of Biology, Molecular Microbiology, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
31
|
Valperga G, de Bono M. Impairing one sensory modality enhances another by reconfiguring peptidergic signalling in Caenorhabditis elegans. eLife 2022; 11:68040. [PMID: 35201977 PMCID: PMC8871372 DOI: 10.7554/elife.68040] [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: 03/03/2021] [Accepted: 02/07/2022] [Indexed: 12/05/2022] Open
Abstract
Animals that lose one sensory modality often show augmented responses to other sensory inputs. The mechanisms underpinning this cross-modal plasticity are poorly understood. We probe such mechanisms by performing a forward genetic screen for mutants with enhanced O2 perception in Caenorhabditis elegans. Multiple mutants exhibiting increased O2 responsiveness concomitantly show defects in other sensory responses. One mutant, qui-1, defective in a conserved NACHT/WD40 protein, abolishes pheromone-evoked Ca2+ responses in the ADL pheromone-sensing neurons. At the same time, ADL responsiveness to pre-synaptic input from O2-sensing neurons is heightened in qui-1, and other sensory defective mutants, resulting in enhanced neurosecretion although not increased Ca2+ responses. Expressing qui-1 selectively in ADL rescues both the qui-1 ADL neurosecretory phenotype and enhanced escape from 21% O2. Profiling ADL neurons in qui-1 mutants highlights extensive changes in gene expression, notably of many neuropeptide receptors. We show that elevated ADL expression of the conserved neuropeptide receptor NPR-22 is necessary for enhanced ADL neurosecretion in qui-1 mutants, and is sufficient to confer increased ADL neurosecretion in control animals. Sensory loss can thus confer cross-modal plasticity by changing the peptidergic connectome.
Collapse
Affiliation(s)
- Giulio Valperga
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.,Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Mario de Bono
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.,Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| |
Collapse
|
32
|
A dominant negative variant of RAB5B disrupts maturation of surfactant protein B and surfactant protein C. Proc Natl Acad Sci U S A 2022; 119:2105228119. [PMID: 35121658 PMCID: PMC8832968 DOI: 10.1073/pnas.2105228119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
The Rab5 GTPase functions in early endosome (EE) fusion in the endocytic pathway. Here, we propose that RAB5B also has a noncanonical vesicular fusion function in the regulated secretion pathway that produces mature surfactant proteins SP-B and SP-C in the lung. This function was revealed from investigation of a proband with interstitial lung disease suggestive of a surfactant dysfunction disorder who carried a de novo Asp136His variant in the RAB5B gene. Our modeling in C. elegans provided information on the genetic and cell biological mechanism, and analyses of proband and normal lung biopsies suggested a function for RAB5B and EEs in surfactant protein processing/trafficking. This work indicates that RAB5B p.Asp136His causes a surfactant dysfunction disorder. Pathogenic variants in surfactant proteins SP-B and SP-C cause surfactant deficiency and interstitial lung disease. Surfactant proteins are synthesized as precursors (proSP-B, proSP-C), trafficked, and processed via a vesicular-regulated secretion pathway; however, control of vesicular trafficking events is not fully understood. Through the Undiagnosed Diseases Network, we evaluated a child with interstitial lung disease suggestive of surfactant deficiency. Variants in known surfactant dysfunction disorder genes were not found in trio exome sequencing. Instead, a de novo heterozygous variant in RAB5B was identified in the Ras/Rab GTPases family nucleotide binding domain, p.Asp136His. Functional studies were performed in Caenorhabditis elegans by knocking the proband variant into the conserved position (Asp135) of the ortholog, rab-5. Genetic analysis demonstrated that rab-5[Asp135His] is damaging, producing a strong dominant negative gene product. rab-5[Asp135His] heterozygotes were also defective in endocytosis and early endosome (EE) fusion. Immunostaining studies of the proband’s lung biopsy revealed that RAB5B and EE marker EEA1 were significantly reduced in alveolar type II cells and that mature SP-B and SP-C were significantly reduced, while proSP-B and proSP-C were normal. Furthermore, staining normal lung showed colocalization of RAB5B and EEA1 with proSP-B and proSP-C. These findings indicate that dominant negative–acting RAB5B Asp136His and EE dysfunction cause a defect in processing/trafficking to produce mature SP-B and SP-C, resulting in interstitial lung disease, and that RAB5B and EEs normally function in the surfactant secretion pathway. Together, the data suggest a noncanonical function for RAB5B and identify RAB5B p.Asp136His as a genetic mechanism for a surfactant dysfunction disorder.
Collapse
|
33
|
Bülow HE. Imaging Glycosaminoglycan Modification Patterns In Vivo. Methods Mol Biol 2022; 2303:539-557. [PMID: 34626406 DOI: 10.1007/978-1-0716-1398-6_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Glycosaminoglycans (GAGs) such as heparan sulfates (HS) or chondroitin sulfates (CS) are long unbranched polymers of a disaccharide comprised of hexuronic acid and hexosamine. Attached to a protein backbone via a characteristic tetrasaccharide, the GAG chains are non-uniformly modified by sulfations, epimerizations, and deacetylations. The resultant glycan chains contain highly modified domains, separated by sections of sparse or no modifications. These GAG domains are central to the role of glycans in binding to proteins and mediating protein-protein interactions. Since HS and CS domains are not genetically encoded, they cannot be visualized and studied with conventional methods in vivo. We describe a transgenic approach using single chain variable fragment (scFv) antibodies that bind HS or CS. By transgenically expressing fluorescently tagged scFv antibodies, we can directly visualize both HS and CS domains in live Caenorhabditis elegans revealing unprecedented cellular specificity and evolutionary conservation (Attreed et al., Nat Methods 9(5): 477-479, 2012; Attreed et al., Glycobiology 26(8): 862-870, 2016) (unpublished). The approach allows concomitant co-labeling of multiple GAG domains, the study of GAG dynamics, and could lend itself to a genetic analysis of GAG domain biosynthesis or function.
Collapse
Affiliation(s)
- Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
34
|
Katz M. Genetic Methods for Cellular Manipulation in C. elegans. Methods Mol Biol 2022; 2468:51-72. [PMID: 35320560 DOI: 10.1007/978-1-0716-2181-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neuron manipulation in vivo by ablation, activation or inactivation, and regulation of gene expression is essential for dissecting nervous system function. Here we describe genetic means for neuron manipulation in the nematode C. elegans, and provide protocols for generating transgenic animals containing these genetic tools.
Collapse
Affiliation(s)
- Menachem Katz
- Department of Biology, Technion- Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
35
|
Wernet N, Wernet V, Fischer R. The small-secreted cysteine-rich protein CyrA is a virulence factor participating in the attack of Caenorhabditis elegans by Duddingtonia flagrans. PLoS Pathog 2021; 17:e1010028. [PMID: 34735554 PMCID: PMC8568293 DOI: 10.1371/journal.ppat.1010028] [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: 06/24/2021] [Accepted: 10/11/2021] [Indexed: 01/14/2023] Open
Abstract
Nematode-trapping fungi (NTF) are a diverse and intriguing group of fungi that live saprotrophically but can switch to a predatory lifestyle when starving and in the presence of nematodes. NTF like Arthrobotrys oligospora or Duddingtonia flagrans produce adhesive trapping networks to catch and immobilize nematodes. After penetration of the cuticle, hyphae grow and develop inside the worm and secrete large amounts of hydrolytic enzymes for digestion. In many microbial pathogenic interactions small-secreted proteins (SSPs) are used to manipulate the host. The genome of D. flagrans encodes more than 100 of such putative SSPs one of which is the cysteine-rich protein CyrA. We have chosen this gene for further analysis because it is only found in NTF and appeared to be upregulated during the interaction. We show that the cyrA gene was transcriptionally induced in trap cells, and the protein accumulated at the inner rim of the hyphal ring before Caenorhabditis elegans capture. After worm penetration, the protein appeared at the fungal infection bulb, where it is likely to be secreted with the help of the exocyst complex. A cyrA-deletion strain was less virulent, and the time from worm capture to paralysis was extended. Heterologous expression of CyrA in C. elegans reduced its lifespan. CyrA accumulated in C. elegans in coelomocytes where the protein possibly is inactivated. This is the first example that SSPs may be important in predatory microbial interactions.
Collapse
Affiliation(s)
- Nicole Wernet
- Karlsruhe Institute of Technology (KIT)—South Campus, Institute for Applied Biosciences, Dept. of Microbiology, Karlsruhe, Germany
| | - Valentin Wernet
- Karlsruhe Institute of Technology (KIT)—South Campus, Institute for Applied Biosciences, Dept. of Microbiology, Karlsruhe, Germany
| | - Reinhard Fischer
- Karlsruhe Institute of Technology (KIT)—South Campus, Institute for Applied Biosciences, Dept. of Microbiology, Karlsruhe, Germany
| |
Collapse
|
36
|
Singh R, Smit RB, Wang X, Wang C, Racher H, Hansen D. Reduction of Derlin activity suppresses Notch-dependent tumours in the C. elegans germ line. PLoS Genet 2021; 17:e1009687. [PMID: 34555015 PMCID: PMC8491880 DOI: 10.1371/journal.pgen.1009687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/05/2021] [Accepted: 09/08/2021] [Indexed: 11/19/2022] Open
Abstract
Regulating the balance between self-renewal (proliferation) and differentiation is key to the long-term functioning of all stem cell pools. In the Caenorhabditis elegans germline, the primary signal controlling this balance is the conserved Notch signaling pathway. Gain-of-function mutations in the GLP-1/Notch receptor cause increased stem cell self-renewal, resulting in a tumour of proliferating germline stem cells. Notch gain-of-function mutations activate the receptor, even in the presence of little or no ligand, and have been associated with many human diseases, including cancers. We demonstrate that reduction in CUP-2 and DER-2 function, which are Derlin family proteins that function in endoplasmic reticulum-associated degradation (ERAD), suppresses the C. elegans germline over-proliferation phenotype associated with glp-1(gain-of-function) mutations. We further demonstrate that their reduction does not suppress other mutations that cause over-proliferation, suggesting that over-proliferation suppression due to loss of Derlin activity is specific to glp-1/Notch (gain-of-function) mutations. Reduction of CUP-2 Derlin activity reduces the expression of a read-out of GLP-1/Notch signaling, suggesting that the suppression of over-proliferation in Derlin loss-of-function mutants is due to a reduction in the activity of the mutated GLP-1/Notch(GF) receptor. Over-proliferation suppression in cup-2 mutants is only seen when the Unfolded Protein Response (UPR) is functioning properly, suggesting that the suppression, and reduction in GLP-1/Notch signaling levels, observed in Derlin mutants may be the result of activation of the UPR. Chemically inducing ER stress also suppress glp-1(gf) over-proliferation but not other mutations that cause over-proliferation. Therefore, ER stress and activation of the UPR may help correct for increased GLP-1/Notch signaling levels, and associated over-proliferation, in the C. elegans germline. Notch signaling is a highly conserved signaling pathway that is utilized in many cell fate decisions in many organisms. In the C. elegans germline, Notch signaling is the primary signal that regulates the balance between stem cell proliferation and differentiation. Notch gain-of-function mutations cause the receptor to be active, even when a signal that is normally needed to activate the receptor is absent. In the germline of C. elegans, gain-of-function mutations in GLP-1, a Notch receptor, results in over-proliferation of the stem cells and tumour formation. Here we demonstrate that a reduction or loss of Derlin activity, which is a conserved family of proteins involved in endoplasmic reticulum-associated degradation (ERAD), suppresses over-proliferation due to GLP-1/Notch gain-of-function mutations. Furthermore, we demonstrate that a surveillance mechanism utilized in cells to monitor and react to proteins that are not folded properly (Unfolded Protein Response-UPR) must be functioning well in order for the loss of Derlin activity to supress over-proliferation caused by glp-1/Notch gain-of-function mutations. This suggests that activation of the UPR may be the mechanism at work for suppressing this type of over-proliferation, when Derlin activity is reduced. Therefore, decreasing Derlin activity may be a means of reducing the impact of phenotypes and diseases due to certain Notch gain-of-function mutations.
Collapse
Affiliation(s)
- Ramya Singh
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Ryan B. Smit
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Xin Wang
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Chris Wang
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Hilary Racher
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Dave Hansen
- Department of Biological Sciences, University of Calgary, Calgary, Canada
- * E-mail:
| |
Collapse
|
37
|
Lukácsi S, Farkas Z, Saskői É, Bajtay Z, Takács-Vellai K. Conserved and Distinct Elements of Phagocytosis in Human and C. elegans. Int J Mol Sci 2021; 22:ijms22168934. [PMID: 34445642 PMCID: PMC8396242 DOI: 10.3390/ijms22168934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Endocytosis provides the cellular nutrition and homeostasis of organisms, but pathogens often take advantage of this entry point to infect host cells. This is counteracted by phagocytosis that plays a key role in the protection against invading microbes both during the initial engulfment of pathogens and in the clearance of infected cells. Phagocytic cells balance two vital functions: preventing the accumulation of cell corpses to avoid pathological inflammation and autoimmunity, whilst maintaining host defence. In this review, we compare elements of phagocytosis in mammals and the nematode Caenorhabditis elegans. Initial recognition of infection requires different mechanisms. In mammals, pattern recognition receptors bind pathogens directly, whereas activation of the innate immune response in the nematode rather relies on the detection of cellular damage. In contrast, molecules involved in efferocytosis—the engulfment and elimination of dying cells and cell debris—are highly conserved between the two species. Therefore, C. elegans is a powerful model to research mechanisms of the phagocytic machinery. Finally, we show that both mammalian and worm studies help to understand how the two phagocytic functions are interconnected: emerging data suggest the activation of innate immunity as a consequence of defective apoptotic cell clearance.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Éva Saskői
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
- Department of Immunology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary
| | - Krisztina Takács-Vellai
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
- Correspondence:
| |
Collapse
|
38
|
Kay RR. Macropinocytosis: Biology and mechanisms. Cells Dev 2021; 168:203713. [PMID: 34175511 DOI: 10.1016/j.cdev.2021.203713] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022]
Abstract
Macropinocytosis is a form of endocytosis performed by ruffles and cups of the plasma membrane. These close to entrap droplets of medium into micron-sized vesicles, which are trafficked through the endocytic system, their contents digested and useful products absorbed. Macropinocytosis is constitutive in certain immune cells and stimulated in many other cells by growth factors. It occurs across the animal kingdom and in amoebae, implying a deep evolutionary history. Its scientific history goes back 100 years, but increasingly work is focused on its medical importance in the immune system, cancer cell feeding, and as a backdoor into cells for viruses and drugs. Macropinocytosis is driven by the actin cytoskeleton whose dynamics can be appreciated with lattice light sheet microscopy: this reveals a surprising variety of routes for forming macropinosomes. In Dictyostelium amoebae, macropinocytic cups are organized around domains of PIP3 and active Ras and Rac in the plasma membrane. These attract activators of the Arp2/3 complex to their periphery, creating rings of actin polymerization that shape the cups. The size of PIP3 domains is controlled by RasGAPs, such as NF1, and the lipid phosphatase, PTEN. It is likely that domain dynamics determine the shape, evolution and closing of macropinocytic structures.
Collapse
Affiliation(s)
- Robert R Kay
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
39
|
Xu Y, Zhang L, Liu Y, Topalidou I, Hassinan C, Ailion M, Zhao Z, Wang T, Chen Z, Bai J. Dopamine receptor DOP-1 engages a sleep pathway to modulate swimming in C. elegans. iScience 2021; 24:102247. [PMID: 33796839 PMCID: PMC7995527 DOI: 10.1016/j.isci.2021.102247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/06/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
Animals require robust yet flexible programs to support locomotion. Here we report a pathway that connects the D1-like dopamine receptor DOP-1 with a sleep mechanism to modulate swimming in C. elegans. We show that DOP-1 plays a negative role in sustaining swimming behavior. By contrast, a pathway through the D2-like dopamine receptor DOP-3 negatively regulates the initiation of swimming, but its impact fades quickly over a few minutes. We find that DOP-1 and the GPCR kinase (G-protein-coupled receptor kinase-2) function in the sleep interneuron RIS, where DOP-1 modulates the secretion of a sleep neuropeptide FLP-11. We further show that DOP-1 and FLP-11 act in the same pathway to modulate swimming. Together, these results delineate a functional connection between a dopamine receptor and a sleep program to regulate swimming in C. elegans. The temporal transition between DOP-3 and DOP-1 pathways highlights the dynamic nature of neuromodulation for rhythmic movements that persist over time.
Collapse
Affiliation(s)
- Ye Xu
- Department of Clinical Medicine, Nanjing Medical University, Nanjing 211166, P. R. China.,Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., Seattle, WA 98109, USA.,Department of Neurology, First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan 570102, P. R. China
| | - Lin Zhang
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., Seattle, WA 98109, USA
| | - Yan Liu
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., Seattle, WA 98109, USA
| | - Irini Topalidou
- Department of Biochemistry, University of Washington, WA 98195
| | - Cera Hassinan
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., Seattle, WA 98109, USA.,Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98019
| | - Michael Ailion
- Department of Biochemistry, University of Washington, WA 98195
| | - Zhenqiang Zhao
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan 570102, P. R. China
| | - Tan Wang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan 570102, P. R. China
| | - Zhibin Chen
- Department of Clinical Medicine, Nanjing Medical University, Nanjing 211166, P. R. China.,Department of Neurology, First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan 570102, P. R. China
| | - Jihong Bai
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., Seattle, WA 98109, USA.,Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98019.,Department of Biochemistry, University of Washington, WA 98195
| |
Collapse
|
40
|
Plagens RN, Mossiah I, Kim Guisbert KS, Guisbert E. Chronic temperature stress inhibits reproduction and disrupts endocytosis via chaperone titration in Caenorhabditis elegans. BMC Biol 2021; 19:75. [PMID: 33858388 PMCID: PMC8051109 DOI: 10.1186/s12915-021-01008-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/19/2021] [Indexed: 11/24/2022] Open
Abstract
Background Temperature influences biology at all levels, from altering rates of biochemical reactions to determining sustainability of entire ecosystems. Although extended exposure to elevated temperatures influences organismal phenotypes important for human health, agriculture, and ecology, the molecular mechanisms that drive these responses remain largely unexplored. Prolonged, mild temperature stress (48 h at 28 °C) has been shown to inhibit reproduction in Caenorhabditis elegans without significantly impacting motility or viability. Results Analysis of molecular responses to chronic stress using RNA-seq uncovers dramatic effects on the transcriptome that are fundamentally distinct from the well-characterized, acute heat shock response (HSR). While a large portion of the genome is differentially expressed ≥ 4-fold after 48 h at 28 °C, the only major class of oogenesis-associated genes affected is the vitellogenin gene family that encodes for yolk proteins (YPs). Whereas YP mRNAs decrease, the proteins accumulate and mislocalize in the pseudocoelomic space as early as 6 h, well before reproduction declines. A trafficking defect in a second, unrelated fluorescent reporter and a decrease in pre-synaptic neuronal signaling indicate that the YP mislocalization is caused by a generalized defect in endocytosis. Molecular chaperones are involved in both endocytosis and refolding damaged proteins. Decreasing levels of the major HSP70 chaperone, HSP-1, causes similar YP trafficking defects in the absence of stress. Conversely, increasing chaperone levels through overexpression of the transcription factor HSF-1 rescues YP trafficking and restores neuronal signaling. Conclusions These data implicate chaperone titration during chronic stress as a molecular mechanism contributing to endocytic defects that influence multiple aspects of organismal physiology. Notably, HSF-1 overexpression improves recovery of viable offspring after exposure to stress. These findings provide important molecular insights into understanding organismal responses to temperature stress as well as phenotypes associated with chronic protein misfolding. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01008-1.
Collapse
Affiliation(s)
- Rosemary N Plagens
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Isiah Mossiah
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Karen S Kim Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Eric Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA.
| |
Collapse
|
41
|
Chaya T, Patel S, Smith EM, Lam A, Miller EN, Clupper M, Kervin K, Tanis JE. A C. elegans genome-wide RNAi screen for altered levamisole sensitivity identifies genes required for muscle function. G3-GENES GENOMES GENETICS 2021; 11:6169532. [PMID: 33713125 PMCID: PMC8049432 DOI: 10.1093/g3journal/jkab047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/09/2021] [Indexed: 01/17/2023]
Abstract
At the neuromuscular junction (NMJ), postsynaptic ionotropic acetylcholine receptors (AChRs) transduce a chemical signal released from a cholinergic motor neuron into an electrical signal to induce muscle contraction. To identify regulators of postsynaptic function, we conducted a genome-wide RNAi screen for genes required for proper response to levamisole, a pharmacological agonist of ionotropic L-AChRs at the Caenorhabditis elegans NMJ. A total of 117 gene knockdowns were found to cause levamisole hypersensitivity, while 18 resulted in levamisole resistance. Our screen identified conserved genes important for muscle function including some that are mutated in congenital myasthenic syndrome, congenital muscular dystrophy, congenital myopathy, myotonic dystrophy, and mitochondrial myopathy. Of the genes found in the screen, we further investigated those predicted to play a role in endocytosis of cell surface receptors. Loss of the Epsin homolog epn-1 caused levamisole hypersensitivity and had opposing effects on the levels of postsynaptic L-AChRs and GABAA receptors, resulting in increased and decreased abundance, respectively. We also examined other genes that resulted in a levamisole-hypersensitive phenotype when knocked down including gas-1, which functions in Complex I of the mitochondrial electron transport chain. Consistent with altered ATP synthesis impacting levamisole response, treatment of wild-type animals with levamisole resulted in L-AChR–dependent depletion of ATP levels. These results suggest that the paralytic effects of levamisole ultimately lead to metabolic exhaustion.
Collapse
Affiliation(s)
- Timothy Chaya
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shrey Patel
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Erin M Smith
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andy Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elaine N Miller
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Michael Clupper
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Kirsten Kervin
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Jessica E Tanis
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
42
|
Serra ND, Sundaram MV. Transcytosis in the development and morphogenesis of epithelial tissues. EMBO J 2021; 40:e106163. [PMID: 33792936 DOI: 10.15252/embj.2020106163] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/21/2020] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Transcytosis is a form of specialized transport through which an extracellular cargo is endocytosed, shuttled across the cytoplasm in membrane-bound vesicles, and secreted at a different plasma membrane surface. This important process allows membrane-impermeable macromolecules to pass through a cell and become accessible to adjacent cells and tissue compartments. Transcytosis also promotes redistribution of plasma membrane proteins and lipids to different regions of the cell surface. Here we review transcytosis and highlight in vivo studies showing how developing epithelial cells use it to change shape, to migrate, and to relocalize signaling molecules.
Collapse
Affiliation(s)
- Nicholas D Serra
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meera V Sundaram
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
43
|
Liu YJ, McIntyre RL, Janssens GE, Williams EG, Lan J, van Weeghel M, Schomakers B, van der Veen H, van der Wel NN, Yao P, Mair WB, Aebersold R, MacInnes AW, Houtkooper RH. Mitochondrial translation and dynamics synergistically extend lifespan in C. elegans through HLH-30. J Cell Biol 2021; 219:151623. [PMID: 32259199 PMCID: PMC7265311 DOI: 10.1083/jcb.201907067] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 01/18/2023] Open
Abstract
Mitochondrial form and function are closely interlinked in homeostasis and aging. Inhibiting mitochondrial translation is known to increase lifespan in C. elegans, and is accompanied by a fragmented mitochondrial network. However, whether this link between mitochondrial translation and morphology is causal in longevity remains uncharacterized. Here, we show in C. elegans that disrupting mitochondrial network homeostasis by blocking fission or fusion synergizes with reduced mitochondrial translation to prolong lifespan and stimulate stress response such as the mitochondrial unfolded protein response, UPRMT. Conversely, immobilizing the mitochondrial network through a simultaneous disruption of fission and fusion abrogates the lifespan increase induced by mitochondrial translation inhibition. Furthermore, we find that the synergistic effect of inhibiting both mitochondrial translation and dynamics on lifespan, despite stimulating UPRMT, does not require it. Instead, this lifespan-extending synergy is exclusively dependent on the lysosome biogenesis and autophagy transcription factor HLH-30/TFEB. Altogether, our study reveals the mechanistic crosstalk between mitochondrial translation, mitochondrial dynamics, and lysosomal signaling in regulating longevity.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Evan G Williams
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology in Zurich, Zurich, Switzerland
| | - Jiayi Lan
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology in Zurich, Zurich, Switzerland
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Bauke Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Henk van der Veen
- Electron Microscopy Center Amsterdam, Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole N van der Wel
- Electron Microscopy Center Amsterdam, Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Pallas Yao
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | - William B Mair
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology in Zurich, Zurich, Switzerland.,Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Alyson W MacInnes
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Tang B, Williams PL, Xue KS, Wang JS, Tang L. Detoxification mechanisms of nickel sulfate in nematode Caenorhabditis elegans. CHEMOSPHERE 2020; 260:127627. [PMID: 32673864 DOI: 10.1016/j.chemosphere.2020.127627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 05/19/2023]
Abstract
Nickel is the most prevailing metal allergen with the highest sensitization rate among the "TOP 25" contact allergens and can affect about 15% of the human population. It is an essential trace metal in plants, animals, and humans. However, the environmental levels of nickel are considerably higher than what is needed for human life. Exposure to high levels of nickel can lead to skin allergies, lung fibrosis, and carcinogenesis. Few existing studies have closely examined the toxicity of nickel, let alone investigated the effective detoxification pathways. Here, we developed a high-throughput screening platform to comprehensively evaluate the nickel toxicity in wild-type C. elegans and explore the underlying detoxification mechanisms in transgenic nematodes. We demonstrated that nickel exerted multiple toxic effects on growth, brood size, feeding, and locomotion in C. elegans. Of which, brood size is the most sensitive endpoint. Nickel was found to first bind to phytochelatin (PC) after entering the worms' body and this PC-Ni complex was further transported by the ABC transporter, CeHMT-1, into the coelomocytes for further detoxification. Our study also demonstrated that the high-throughput screening platform is a promising system for evaluation and investigation of the ecological risks of heavy metals.
Collapse
Affiliation(s)
- Bowen Tang
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, USA
| | - Phillip L Williams
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, USA
| | - Kathy S Xue
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, USA
| | - Jia-Sheng Wang
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, USA
| | - Lili Tang
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, USA.
| |
Collapse
|
45
|
Affiliation(s)
- Brant M Webster
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Holly K Gildea
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Andrew Dillin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA. .,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA. .,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
46
|
Walsh MB, Janzen E, Wingrove E, Hosseinibarkooie S, Muela NR, Davidow L, Dimitriadi M, Norabuena EM, Rubin LL, Wirth B, Hart AC. Genetic modifiers ameliorate endocytic and neuromuscular defects in a model of spinal muscular atrophy. BMC Biol 2020; 18:127. [PMID: 32938453 PMCID: PMC7495824 DOI: 10.1186/s12915-020-00845-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/11/2020] [Indexed: 12/31/2022] Open
Abstract
Background Understanding the genetic modifiers of neurodegenerative diseases can provide insight into the mechanisms underlying these disorders. Here, we examine the relationship between the motor neuron disease spinal muscular atrophy (SMA), which is caused by reduced levels of the survival of motor neuron (SMN) protein, and the actin-bundling protein Plastin 3 (PLS3). Increased PLS3 levels suppress symptoms in a subset of SMA patients and ameliorate defects in SMA disease models, but the functional connection between PLS3 and SMN is poorly understood. Results We provide immunohistochemical and biochemical evidence for large protein complexes localized in vertebrate motor neuron processes that contain PLS3, SMN, and members of the hnRNP F/H family of proteins. Using a Caenorhabditis elegans (C. elegans) SMA model, we determine that overexpression of PLS3 or loss of the C. elegans hnRNP F/H ortholog SYM-2 enhances endocytic function and ameliorates neuromuscular defects caused by decreased SMN-1 levels. Furthermore, either increasing PLS3 or decreasing SYM-2 levels suppresses defects in a C. elegans ALS model. Conclusions We propose that hnRNP F/H act in the same protein complex as PLS3 and SMN and that the function of this complex is critical for endocytic pathways, suggesting that hnRNP F/H proteins could be potential targets for therapy development.
Collapse
Affiliation(s)
- Melissa B Walsh
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA
| | - Eva Janzen
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Emily Wingrove
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA
| | - Seyyedmohsen Hosseinibarkooie
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Natalia Rodriguez Muela
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Lance Davidow
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Maria Dimitriadi
- Department of Biological and Environmental Sciences, University of Hertfordshire, Hertfordshire, UK
| | - Erika M Norabuena
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Anne C Hart
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA.
| |
Collapse
|
47
|
Lin XP, Mintern JD, Gleeson PA. Macropinocytosis in Different Cell Types: Similarities and Differences. MEMBRANES 2020; 10:membranes10080177. [PMID: 32756454 PMCID: PMC7463864 DOI: 10.3390/membranes10080177] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Abstract
Macropinocytosis is a unique pathway of endocytosis characterised by the nonspecific internalisation of large amounts of extracellular fluid, solutes and membrane in large endocytic vesicles known as macropinosomes. Macropinocytosis is important in a range of physiological processes, including antigen presentation, nutrient sensing, recycling of plasma proteins, migration and signalling. It has become apparent in recent years from the study of specialised cells that there are multiple pathways of macropinocytosis utilised by different cell types, and some of these pathways are triggered by different stimuli. Understanding the physiological function of macropinocytosis requires knowledge of the regulation and fate of the macropinocytosis pathways in a range of cell types. Here, we compare the mechanisms of macropinocytosis in different primary and immortalised cells, identify the gaps in knowledge in the field and discuss the potential approaches to analyse the function of macropinocytosis in vivo.
Collapse
|
48
|
Extracellular proteostasis prevents aggregation during pathogenic attack. Nature 2020; 584:410-414. [PMID: 32641833 DOI: 10.1038/s41586-020-2461-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/16/2020] [Indexed: 12/26/2022]
Abstract
In metazoans, the secreted proteome participates in intercellular signalling and innate immunity, and builds the extracellular matrix scaffold around cells. Compared with the relatively constant intracellular environment, conditions for proteins in the extracellular space are harsher, and low concentrations of ATP prevent the activity of intracellular components of the protein quality-control machinery. Until now, only a few bona fide extracellular chaperones and proteases have been shown to limit the aggregation of extracellular proteins1-5. Here we performed a systematic analysis of the extracellular proteostasis network in Caenorhabditis elegans with an RNA interference screen that targets genes that encode the secreted proteome. We discovered 57 regulators of extracellular protein aggregation, including several proteins related to innate immunity. Because intracellular proteostasis is upregulated in response to pathogens6-9, we investigated whether pathogens also stimulate extracellular proteostasis. Using a pore-forming toxin to mimic a pathogenic attack, we found that C. elegans responded by increasing the expression of components of extracellular proteostasis and by limiting aggregation of extracellular proteins. The activation of extracellular proteostasis was dependent on stress-activated MAP kinase signalling. Notably, the overexpression of components of extracellular proteostasis delayed ageing and rendered worms resistant to intoxication. We propose that enhanced extracellular proteostasis contributes to systemic host defence by maintaining a functional secreted proteome and avoiding proteotoxicity.
Collapse
|
49
|
A secreted microRNA disrupts autophagy in distinct tissues of Caenorhabditis elegans upon ageing. Nat Commun 2019; 10:4827. [PMID: 31645592 PMCID: PMC6811558 DOI: 10.1038/s41467-019-12821-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 09/05/2019] [Indexed: 12/25/2022] Open
Abstract
Macroautophagy, a key player in protein quality control, is proposed to be systematically impaired in distinct tissues and causes coordinated disruption of protein homeostasis and ageing throughout the body. Although tissue-specific changes in autophagy and ageing have been extensively explored, the mechanism underlying the inter-tissue regulation of autophagy with ageing is poorly understood. Here, we show that a secreted microRNA, mir-83/miR-29, controls the age-related decrease in macroautophagy across tissues in Caenorhabditis elegans. Upregulated in the intestine by hsf-1/HSF1 with age, mir-83 is transported across tissues potentially via extracellular vesicles and disrupts macroautophagy by suppressing CUP-5/MCOLN, a vital autophagy regulator, autonomously in the intestine as well as non-autonomously in body wall muscle. Mutating mir-83 thereby enhances macroautophagy in different tissues, promoting protein homeostasis and longevity. These findings thus identify a microRNA-based mechanism to coordinate the decreasing macroautophagy in various tissues with age. Decreased autophagy is a hallmark of ageing, but its inter-tissue regulation is poorly understood. Here, Zhou et al. identify mir-83 in C. elegans, which is transported across tissues and suppresses autophagy, contributing to age-related decline.
Collapse
|
50
|
Eom HJ, Choi J. Clathrin-mediated endocytosis is involved in uptake and toxicity of silica nanoparticles in Caenohabditis elegans. Chem Biol Interact 2019; 311:108774. [DOI: 10.1016/j.cbi.2019.108774] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022]
|