101
|
Mahapatra A, Dhakal A, Noguchi A, Vadlamani P, Hundley HA. ADAR-mediated regulation of PQM-1 expression in neurons impacts gene expression throughout C. elegans and regulates survival from hypoxia. PLoS Biol 2023; 21:e3002150. [PMID: 37747897 PMCID: PMC10553819 DOI: 10.1371/journal.pbio.3002150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/05/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
The ability to alter gene expression programs in response to changes in environmental conditions is central to the ability of an organism to thrive. For most organisms, the nervous system serves as the master regulator in communicating information about the animal's surroundings to other tissues. The information relay centers on signaling pathways that cue transcription factors in a given cell type to execute a specific gene expression program, but also provide a means to signal between tissues. The transcription factor PQM-1 is an important mediator of the insulin signaling pathway contributing to longevity and the stress response as well as impacting survival from hypoxia. Herein, we reveal a novel mechanism for regulating PQM-1 expression specifically in neural cells of larval animals. Our studies reveal that the RNA-binding protein (RBP), ADR-1, binds to pqm-1 mRNA in neural cells. This binding is regulated by the presence of a second RBP, ADR-2, which when absent leads to reduced expression of both pqm-1 and downstream PQM-1 activated genes. Interestingly, we find that neural pqm-1 expression is sufficient to impact gene expression throughout the animal and affect survival from hypoxia, phenotypes that we also observe in adr mutant animals. Together, these studies reveal an important posttranscriptional gene regulatory mechanism in Caenorhabditis elegans that allows the nervous system to sense and respond to environmental conditions to promote organismal survival from hypoxia.
Collapse
Affiliation(s)
- Ananya Mahapatra
- Genome, Cell and Developmental Biology Graduate Program, Indiana University, Bloomington, Indiana, United States of America
| | - Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine–Bloomington, Bloomington, Indiana, United States of America
| | - Aika Noguchi
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Pranathi Vadlamani
- Medical Sciences Program, Indiana University School of Medicine–Bloomington, Bloomington, Indiana, United States of America
| | - Heather A. Hundley
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
102
|
Shiri TJ, Viau C, Gu X, Xu L, Lu Y, Xia J. The Native Microbiome Member Chryseobacterium sp. CHNTR56 MYb120 Induces Trehalose Production via a Shift in Central Carbon Metabolism during Early Life in C. elegans. Metabolites 2023; 13:953. [PMID: 37623896 PMCID: PMC10456584 DOI: 10.3390/metabo13080953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
Aging is the system-wide loss of homeostasis, eventually leading to death. There is growing evidence that the microbiome not only evolves with its aging host, but also directly affects aging via the modulation of metabolites involved in important cellular functions. The widely used model organism C. elegans exhibits high selectivity towards its native microbiome members which confer a range of differential phenotypes and possess varying functional capacities. The ability of one such native microbiome species, Chryseobacterium sp. CHNTR56 MYb120, to improve the lifespan of C. elegans and to promote the production of Vitamin B6 in the co-colonizing member Comamonas sp. 12022 MYb131 are some of its beneficial effects on the worm host. We hypothesize that studying its metabolic influence on the different life stages of the worm could provide further insights into mutualistic interactions. The present work applied LC-MS untargeted metabolomics and isotope labeling to study the impact of the native microbiome member Chryseobacterium sp. CHNTR56 MYb120 on the metabolism of C. elegans. In addition to the upregulation of biosynthesis and detoxification pathway intermediates, we found that Chryseobacterium sp. CHNTR56 MYb120 upregulates the glyoxylate shunt in mid-adult worms which is linked to the upregulation of trehalose, an important metabolite for desiccation tolerance in older worms.
Collapse
Affiliation(s)
- Tanisha Jean Shiri
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (T.J.S.); (C.V.); (X.G.); (L.X.)
| | - Charles Viau
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (T.J.S.); (C.V.); (X.G.); (L.X.)
| | - Xue Gu
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (T.J.S.); (C.V.); (X.G.); (L.X.)
| | - Lei Xu
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (T.J.S.); (C.V.); (X.G.); (L.X.)
| | - Yao Lu
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada;
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (T.J.S.); (C.V.); (X.G.); (L.X.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada;
| |
Collapse
|
103
|
Tsai YT, Chang CH, Tsai HY. Rege-1 promotes C. elegans survival by modulating IIS and TOR pathways. PLoS Genet 2023; 19:e1010869. [PMID: 37556491 PMCID: PMC10441803 DOI: 10.1371/journal.pgen.1010869] [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: 11/29/2022] [Revised: 08/21/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
Metabolic pathways are known to sense the environmental stimuli and result in physiological adjustments. The responding processes need to be tightly controlled. Here, we show that upon encountering P. aeruginosa, C. elegans upregulate the transcription factor ets-4, but this upregulation is attenuated by the ribonuclease, rege-1. As such, mutants with defective REGE-1 ribonuclease activity undergo ets-4-dependent early death upon challenge with P. aeruginosa. Furthermore, mRNA-seq analysis revealed associated global changes in two key metabolic pathways, the IIS (insulin/IGF signaling) and TOR (target of rapamycin) kinase signaling pathways. In particular, failure to degrade ets-4 mRNA in activity-defective rege-1 mutants resulted in upregulation of class II longevity genes, which are suppressed during longevity, and activation of TORC1 kinase signaling pathway. Genetic inhibition of either pathway way was sufficient to abolish the poor survival phenotype in rege-1 worms. Further analysis of ETS-4 ChIP data from ENCODE and characterization of one upregulated class II gene, ins-7, support that the Class II genes are activated by ETS-4. Interestingly, deleting an upregulated Class II gene, acox-1.5, a peroxisome β-oxidation enzyme, largely rescues the fat lost phenotype and survival difference between rege-1 mutants and wild-types. Thus, rege-1 appears to be crucial for animal survival due to its tight regulation of physiological responses to environmental stimuli. This function is reminiscent of its mammalian ortholog, Regnase-1, which modulates the intestinal mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Hsi Chang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yue Tsai
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
104
|
Egan BM, Pohl F, Anderson X, Williams SC, Adodo IG, Hunt P, Wang Z, Chiu CH, Scharf A, Mosley M, Kumar S, Schneider DL, Fujiwara H, Hsu FF, Kornfeld K. The ACE-inhibitor drug captopril inhibits ACN-1 to control dauer formation and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.17.549402. [PMID: 37502959 PMCID: PMC10370070 DOI: 10.1101/2023.07.17.549402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The renin-angiotensin-aldosterone system (RAAS) plays a well-characterized role regulating blood pressure in mammals. Pharmacological and genetic manipulation of the RAAS has been shown to extend lifespan in C. elegans , Drosophila , and rodents, but its mechanism is not well defined. Here we investigate the angiotensin-converting enzyme (ACE) inhibitor drug captopril, which extends lifespan in worms and mice. To investigate the mechanism, we performed a forward genetic screen for captopril hypersensitive mutants. We identified a missense mutation that causes a partial loss-of-function of the daf-2 receptor tyrosine kinase gene, a powerful regulator of aging. The homologous mutation in the human insulin receptor causes Donohue syndrome, establishing these mutant worms as an invertebrate model of this disease. Captopril functions in C. elegans by inhibiting ACN-1, the worm homolog of ACE. Reducing the activity of acn-1 via captopril or RNAi promoted dauer larvae formation, suggesting acn-1 is a daf gene. Captopril-mediated lifespan extension xwas abrogated by daf-16(lf) and daf-12(lf) mutations. Our results indicate that captopril and acn-1 control aging by modulating dauer formation pathways. We speculate that this represents a conserved mechanism of lifespan control. Summary Statement Captopril and acn-1 control aging. By demonstrating they regulate dauer formation and interact with daf genes, including a new DAF-2(A261V) mutant corresponding to a human disease variant, we clarified the mechanism.
Collapse
|
105
|
Jacob PL, Brugnoli B, Del Giudice A, Phan H, Chauhan VM, Beckett L, Gillis RB, Moloney C, Cavanagh RJ, Krumins E, Reynolds-Green M, Lentz JC, Conte C, Cuzzucoli Crucitti V, Couturaud B, Galantini L, Francolini I, Howdle SM, Taresco V. Poly (diglycerol adipate) variants as enhanced nanocarrier replacements in drug delivery applications. J Colloid Interface Sci 2023; 641:1043-1057. [PMID: 36996683 DOI: 10.1016/j.jcis.2023.03.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023]
Abstract
Sustainably derived poly(glycerol adipate) (PGA) has been deemed to deliver all the desirable features expected in a polymeric scaffold for drug-delivery, including biodegradability, biocompatibility, self-assembly into nanoparticles (NPs) and a functionalisable pendant group. Despite showing these advantages over commercial alkyl polyesters, PGA suffers from a series of key drawbacks caused by poor amphiphilic balance. This leads to weak drug-polymer interactions and subsequent low drug-loading in NPs, as well as low NPs stability. To overcome this, in the present work, we applied a more significant variation of the polyester backbone while maintaining mild and sustainable polymerisation conditions. We have investigated the effect of the variation of both hydrophilic and hydrophobic segments upon physical properties and drug interactions as well as self-assembly and NPs stability. For the first time we have replaced glycerol with the more hydrophilic diglycerol, as well as adjusting the final amphiphilic balance of the polyester repetitive units by incorporating the more hydrophobic 1,6-n-hexanediol (Hex). The properties of the novel poly(diglycerol adipate) (PDGA) variants have been compared against known polyglycerol-based polyesters. Interestingly, while the bare PDGA showed improved water solubility and diminished self-assembling ability, the Hex variation demonstrated enhanced features as a nanocarrier. In this regard, PDGAHex NPs were tested for their stability in different environments and for their ability to encode enhanced drug loading. Moreover, the novel materials have shown good biocompatibility in both in vitro and in vivo (whole organism) experiments.
Collapse
Affiliation(s)
- Philippa L Jacob
- School of Chemistry, University Park, Nottingham NG7 2RD, United Kingdom
| | - Benedetta Brugnoli
- Dept. of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | | | - Hien Phan
- Institut de Chimie et des Matériaux Paris-Est, Université de Paris-Est Créteil, CNRS UMR 7182, 2 rue Henri Dunant, 94320 Thiais, France
| | - Veeren M Chauhan
- Laboratory of Biophysics and Surface Analysis, School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham NG7 2RD, United Kingdom
| | - Laura Beckett
- Laboratory of Biophysics and Surface Analysis, School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham NG7 2RD, United Kingdom
| | - Richard B Gillis
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington LE12 5RD, United Kingdom; Biomaterials Group, School of Biosciences, University of Nottingham, Sutton Bonington LE12 5RD, United Kingdom; College of Business, Technology and Engineering, Sheffield Hallam University, Food and Nutrition Group, Sheffield S1 1WB, United Kingdom
| | - Cara Moloney
- School of Medicine, BioDiscovery Institute-3, University Park, Nottingham NG7 2RD, United Kingdom
| | - Robert J Cavanagh
- School of Medicine, BioDiscovery Institute-3, University Park, Nottingham NG7 2RD, United Kingdom
| | - Eduards Krumins
- School of Chemistry, University Park, Nottingham NG7 2RD, United Kingdom
| | | | - Joachim C Lentz
- School of Chemistry, University Park, Nottingham NG7 2RD, United Kingdom
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Valentina Cuzzucoli Crucitti
- Centre for Additive Manufacturing and Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Benoit Couturaud
- Institut de Chimie et des Matériaux Paris-Est, Université de Paris-Est Créteil, CNRS UMR 7182, 2 rue Henri Dunant, 94320 Thiais, France
| | - Luciano Galantini
- Dept. of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Iolanda Francolini
- Dept. of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Steven M Howdle
- School of Chemistry, University Park, Nottingham NG7 2RD, United Kingdom
| | - Vincenzo Taresco
- School of Chemistry, University Park, Nottingham NG7 2RD, United Kingdom.
| |
Collapse
|
106
|
Soo SK, Rudich ZD, Ko B, Moldakozhayev A, AlOkda A, Van Raamsdonk JM. Biological resilience and aging: Activation of stress response pathways contributes to lifespan extension. Ageing Res Rev 2023; 88:101941. [PMID: 37127095 DOI: 10.1016/j.arr.2023.101941] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/06/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
While aging was traditionally viewed as a stochastic process of damage accumulation, it is now clear that aging is strongly influenced by genetics. The identification and characterization of long-lived genetic mutants in model organisms has provided insights into the genetic pathways and molecular mechanisms involved in extending longevity. Long-lived genetic mutants exhibit activation of multiple stress response pathways leading to enhanced resistance to exogenous stressors. As a result, lifespan exhibits a significant, positive correlation with resistance to stress. Disruption of stress response pathways inhibits lifespan extension in multiple long-lived mutants representing different pathways of lifespan extension and can also reduce the lifespan of wild-type animals. Combined, this suggests that activation of stress response pathways is a key mechanism by which long-lived mutants achieve their extended longevity and that many of these pathways are also required for normal lifespan. These results highlight an important role for stress response pathways in determining the lifespan of an organism.
Collapse
Affiliation(s)
- Sonja K Soo
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Zenith D Rudich
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Bokang Ko
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alibek Moldakozhayev
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Abdelrahman AlOkda
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
107
|
Fan S, Yan Y, Xia Y, Zhou Z, Luo L, Zhu M, Han Y, Yao D, Zhang L, Fang M, Peng L, Yu J, Liu Y, Gao X, Guan H, Li H, Wang C, Wu X, Zhu H, Cao Y, Huang C. Pregnane X receptor agonist nomilin extends lifespan and healthspan in preclinical models through detoxification functions. Nat Commun 2023; 14:3368. [PMID: 37291126 PMCID: PMC10250385 DOI: 10.1038/s41467-023-39118-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
Citrus fruit has long been considered a healthy food, but its role and detailed mechanism in lifespan extension are not clear. Here, by using the nematode C. elegans, we identified that nomilin, a bitter-taste limoloid that is enriched in citrus, significantly extended the animals' lifespan, healthspan, and toxin resistance. Further analyses indicate that this ageing inhibiting activity depended on the insulin-like pathway DAF-2/DAF-16 and nuclear hormone receptors NHR-8/DAF-12. Moreover, the human pregnane X receptor (hPXR) was identified as the mammalian counterpart of NHR-8/DAF-12 and X-ray crystallography showed that nomilin directly binds with hPXR. The hPXR mutations that prevented nomilin binding blocked the activity of nomilin both in mammalian cells and in C. elegans. Finally, dietary nomilin supplementation improved healthspan and lifespan in D-galactose- and doxorubicin-induced senescent mice as well as in male senescence accelerated mice prone 8 (SAMP8) mice, and induced a longevity gene signature similar to that of most longevity interventions in the liver of bile-duct-ligation male mice. Taken together, we identified that nomilin may extend lifespan and healthspan in animals via the activation of PXR mediated detoxification functions.
Collapse
Affiliation(s)
- Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingxuan Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Xia
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, 200125, China
| | - Zhenyu Zhou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lingling Luo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengnan Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- CAS Center for Excellence in Molecular Cell Science; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongli Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Deqiang Yao
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Lijun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Minglv Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lina Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- CAS Center for Excellence in Molecular Cell Science; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Yu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyan Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongli Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaojun Wu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huanhu Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yu Cao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai, 200125, China.
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
108
|
Sun B, Kim H, Mello CC, Priess JR. The CERV protein of Cer1, a C. elegans LTR retrotransposon, is required for nuclear export of viral genomic RNA and can form giant nuclear rods. PLoS Genet 2023; 19:e1010804. [PMID: 37384599 PMCID: PMC10309623 DOI: 10.1371/journal.pgen.1010804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Retroviruses and closely related LTR retrotransposons export full-length, unspliced genomic RNA (gRNA) for packaging into virions and to serve as the mRNA encoding GAG and POL polyproteins. Because gRNA often includes splice acceptor and donor sequences used to splice viral mRNAs, retroelements must overcome host mechanisms that retain intron-containing RNAs in the nucleus. Here we examine gRNA expression in Cer1, an LTR retrotransposon in C. elegans which somehow avoids silencing and is highly expressed in germ cells. Newly exported Cer1 gRNA associates rapidly with the Cer1 GAG protein, which has structural similarity with retroviral GAG proteins. gRNA export requires CERV (C. elegans regulator of viral expression), a novel protein encoded by a spliced Cer1 mRNA. CERV phosphorylation at S214 is essential for gRNA export, and phosphorylated CERV colocalizes with nuclear gRNA at presumptive sites of transcription. By electron microscopy, tagged CERV proteins surround clusters of distinct, linear fibrils that likely represent gRNA molecules. Single fibrils, or groups of aligned fibrils, also localize near nuclear pores. During the C. elegans self-fertile period, when hermaphrodites fertilize oocytes with their own sperm, CERV concentrates in two nuclear foci that are coincident with gRNA. However, as hermaphrodites cease self-fertilization, and can only produce cross-progeny, CERV undergoes a remarkable transition to form giant nuclear rods or cylinders that can be up to 5 microns in length. We propose a novel mechanism of rod formation, in which stage-specific changes in the nucleolus induce CERV to localize to the nucleolar periphery in flattened streaks of protein and gRNA; these streaks then roll up into cylinders. The rods are a widespread feature of Cer1 in wild strains of C. elegans, but their function is not known and might be limited to cross-progeny. We speculate that the adaptive strategy Cer1 uses for the identical self-progeny of a host hermaphrodite might differ for heterozygous cross-progeny sired by males. For example, mating introduces male chromosomes which can have different, or no, Cer1 elements.
Collapse
Affiliation(s)
- Bing Sun
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester,United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Haram Kim
- Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Craig C. Mello
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester,United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - James R. Priess
- Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
109
|
Shao X, Zhang M, Chen Y, Sun S, Yang S, Li Q. Exosome-mediated delivery of superoxide dismutase for anti-aging studies in Caenorhabditis elegans. Int J Pharm 2023; 641:123090. [PMID: 37268030 DOI: 10.1016/j.ijpharm.2023.123090] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/09/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Aging is a dynamic and progressive process mediated by reactive oxygen species (ROS), and the antioxidant enzyme superoxide dismutase (SOD) can effectively scavenge ROS to extend longevity. However, the instability and impermeability of native enzyme limit its in vivo biomedical application. Currently, exosome as protein carriers attracts considerable attention in the disease treatment owing to low immunogenicity and high stability. Herein, SOD was encapsulated into exosomes via mechanical extrusion with saponin permeabilization to obtain SOD-loaded EXO (SOD@EXO). SOD@EXO with a hydrodynamic diameter of 101.7 ± 5.6 nm could scavenge excessive ROS and protect the cells from oxidative damage induced by 1-methyl-4-phenylpyridine. Compared with native SOD, SOD@EXO significantly extended the lifespan of N2 wild-type Caenorhabditis elegans under normal conditions. Moreover, SOD@EXO improved the resistance against heat and oxidative stress, leading to notable survival ratio under these hostile conditions. Overall, the exosome-mediated delivery of SOD could reduce ROS level and delay aging in C. elegans model, thereby providing potential strategies to treat ROS-related diseases in future.
Collapse
Affiliation(s)
- Xinxin Shao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Mengzhu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yingxuan Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Siyu Sun
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Shengcai Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
110
|
Belavgeni A, Maremonti F, Tonnus W, Stadtmüller M, Gavali S, Mallais M, Flade K, Brucker A, Becker JN, Beer K, Tmava M, Stumpf J, Gembardt F, Hugo C, Giacca M, Hale BG, Perakakis N, Sha W, Pratt DA, Schally AV, Bornstein SR, Linkermann A. vPIF-1 is an insulin-like antiferroptotic viral peptide. Proc Natl Acad Sci U S A 2023; 120:e2300320120. [PMID: 37186845 PMCID: PMC10214148 DOI: 10.1073/pnas.2300320120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Iridoviridae, such as the lymphocystis disease virus-1 (LCDV-1) and other viruses, encode viral insulin-like peptides (VILPs) which are capable of triggering insulin receptors (IRs) and insulin-like growth factor receptors. The homology of VILPs includes highly conserved disulfide bridges. However, the binding affinities to IRs were reported to be 200- to 500-fold less effective compared to the endogenous ligands. We therefore speculated that these peptides also have noninsulin functions. Here, we report that the LCDV-1 VILP can function as a potent and highly specific inhibitor of ferroptosis. Induction of cell death by the ferroptosis inducers erastin, RSL3, FIN56, and FINO2 and nonferroptotic necrosis produced by the thioredoxin-reductase inhibitor ferroptocide were potently prevented by LCDV-1, while human insulin had no effect. Fas-induced apoptosis, necroptosis, mitotane-induced cell death and growth hormone-releasing hormone antagonist-induced necrosis were unaffected, suggesting the specificity to ferroptosis inhibition by the LCDV-1 VILP. Mechanistically, we identified the viral C-peptide to be required for inhibition of lipid peroxidation and ferroptosis inhibition, while the human C-peptide exhibited no antiferroptotic properties. In addition, the deletion of the viral C-peptide abolishes radical trapping activity in cell-free systems. We conclude that iridoviridae, through the expression of insulin-like viral peptides, are capable of preventing ferroptosis. In analogy to the viral mitochondrial inhibitor of apoptosis and the viral inhibitor of RIP activation (vIRA) that prevents necroptosis, we rename the LCDV-1 VILP a viral peptide inhibitor of ferroptosis-1. Finally, our findings indicate that ferroptosis may function as a viral defense mechanism in lower organisms.
Collapse
Affiliation(s)
- Alexia Belavgeni
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Francesca Maremonti
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Marlena Stadtmüller
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Shubhangi Gavali
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Melodie Mallais
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ONK1N 6N5, Canada
| | - Karolin Flade
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Anne Brucker
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Jorunn Naila Becker
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Kristina Beer
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Mirela Tmava
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Julian Stumpf
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Florian Gembardt
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
| | - Mauro Giacca
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, WC2R 2LSLondon, United Kingdom
| | - Benjamin G. Hale
- Institute of Medical Virology, University of Zürich8057, Zürich, Switzerland
| | - Nikolaos Perakakis
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307Dresden, Germany
| | - Wei Sha
- Veterans Affairs Medical Center, Miami, FL33125
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL33150
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL33136
- Division of Medical Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL33136
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL33136
| | - Derek A. Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ONK1N 6N5, Canada
| | - Andrew V. Schally
- Veterans Affairs Medical Center, Miami, FL33125
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL33150
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL33136
- Division of Medical Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL33136
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL33136
| | - Stefan R. Bornstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, WC2R 2LSLondon, United Kingdom
- Center for Regenerative Therapies, Technische Universität Dresden, 01307Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of Technische Universität Dresden, Faculty of Medicine, 01307Dresden, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921Singapore, Singapore
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY10461
| |
Collapse
|
111
|
Jaskolowski M, Jomaa A, Gamerdinger M, Shrestha S, Leibundgut M, Deuerling E, Ban N. Molecular basis of the TRAP complex function in ER protein biogenesis. Nat Struct Mol Biol 2023:10.1038/s41594-023-00990-0. [PMID: 37170030 DOI: 10.1038/s41594-023-00990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/06/2023] [Indexed: 05/13/2023]
Abstract
The translocon-associated protein (TRAP) complex resides in the endoplasmic reticulum (ER) membrane and interacts with the Sec translocon and the ribosome to facilitate biogenesis of secretory and membrane proteins. TRAP plays a key role in the secretion of many hormones, including insulin. Here we reveal the molecular architecture of the mammalian TRAP complex and how it engages the translating ribosome associated with Sec61 translocon on the ER membrane. The TRAP complex is anchored to the ribosome via a long tether and its position is further stabilized by a finger-like loop. This positions a cradle-like lumenal domain of TRAP below the translocon for interactions with translocated nascent chains. Our structure-guided TRAP mutations in Caenorhabditis elegans lead to growth deficits associated with increased ER stress and defects in protein hormone secretion. These findings elucidate the molecular basis of the TRAP complex in the biogenesis and translocation of proteins at the ER.
Collapse
Affiliation(s)
- Mateusz Jaskolowski
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Ahmad Jomaa
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland.
- Department of Molecular Physiology and Biological Physics and the Center for Cell and Membrane Physiology, University of Virginia, Charlottesville, VA, USA.
| | - Martin Gamerdinger
- Department of Biology, Molecular Microbiology, University of Konstanz, Konstanz, Germany
| | - Sandeep Shrestha
- Department of Biology, Molecular Microbiology, University of Konstanz, Konstanz, Germany
| | - Marc Leibundgut
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Elke Deuerling
- Department of Biology, Molecular Microbiology, University of Konstanz, Konstanz, Germany.
| | - Nenad Ban
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
112
|
Mahapatra A, Dhakal A, Noguchi A, Vadlamani P, Hundley HA. ADARs employ a neural-specific mechanism to regulate PQM-1 expression and survival from hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539519. [PMID: 37205482 PMCID: PMC10187282 DOI: 10.1101/2023.05.05.539519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The ability to alter gene expression programs in response to changes in environmental conditions is central to the ability of an organism to thrive. For most organisms, the nervous system serves as the master regulator in communicating information about the animal's surroundings to other tissues. The information relay centers on signaling pathways that cue transcription factors in a given cell type to execute a specific gene expression program, but also provide a means to signal between tissues. The transcription factor PQM-1 is an important mediator of the insulin signaling pathway contributing to longevity and the stress response as well as impacting survival from hypoxia. Herein, we reveal a novel mechanism for regulating PQM-1 expression specifically in neural cells of larval animals. Our studies reveal that the RNA binding protein, ADR-1, binds to pqm-1 mRNA in neural cells. This binding is regulated by the presence of a second RNA binding protein, ADR-2, which when absent leads to reduced expression of both pqm-1 and downstream PQM-1 activated genes. Interestingly, we find that neural pqm-1 expression is sufficient to impact gene expression throughout the animal and affect survival from hypoxia; phenotypes that we also observe in adr mutant animals. Together, these studies reveal an important post-transcriptional gene regulatory mechanism that allows the nervous system to sense and respond to environmental conditions to promote organismal survival from hypoxia.
Collapse
Affiliation(s)
- Ananya Mahapatra
- Genome, Cell and Developmental Biology Graduate Program, Indiana University, Bloomington IN, 47405 USA
| | - Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine – Bloomington, Bloomington IN, 47405 USA
| | - Aika Noguchi
- Department of Biology, Indiana University, Bloomington IN 47405 USA
| | - Pranathi Vadlamani
- Medical Sciences Program, Indiana University School of Medicine – Bloomington, Bloomington IN, 47405 USA
| | | |
Collapse
|
113
|
Ohta A, Yamashiro S, Kuhara A. Temperature acclimation: Temperature shift induces system conversion to cold tolerance in C. elegans. Neurosci Res 2023:S0168-0102(23)00075-5. [PMID: 37086751 DOI: 10.1016/j.neures.2023.04.005] [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: 01/31/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/24/2023]
Abstract
Acclimation to temperature is one of the survival strategies used by organisms to adapt to changing environmental temperatures. Caenorhabditis elegans' cold tolerance is altered by previous cultivation temperature, and similarly, past low-temperature induces a longer lifespan. Temperature is thought to cause a large shift in homeostasis, lipid metabolism, and reproduction in the organism because it is a direct physiological factor during chemical events. This paper will share and discuss what we know so far about the neural and molecular mechanisms that control cold tolerance and lifespan by altering lipid metabolism and physiological characteristics. We hope that this will contribute to a better understanding of how organisms respond to temperature changes.
Collapse
Affiliation(s)
- Akane Ohta
- Graduate School of Natural Science, Konan University, Kobe 658-8501, JAPAN; Faculty of Science and Engineering, Konan University, Kobe 658-8501, JAPAN; Institute for Integrative Neurobiology, Konan University, Kobe 658-8501, JAPAN; AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo 100-0004, JAPAN.
| | - Serina Yamashiro
- Graduate School of Natural Science, Konan University, Kobe 658-8501, JAPAN; Institute for Integrative Neurobiology, Konan University, Kobe 658-8501, JAPAN
| | - Atsushi Kuhara
- Graduate School of Natural Science, Konan University, Kobe 658-8501, JAPAN; Faculty of Science and Engineering, Konan University, Kobe 658-8501, JAPAN; Institute for Integrative Neurobiology, Konan University, Kobe 658-8501, JAPAN; AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo 100-0004, JAPAN.
| |
Collapse
|
114
|
Chen WW, Tang W, Hamerton EK, Kuo PX, Lemieux GA, Ashrafi K, Cicerone MT. Identifying lipid particle sub-types in live Caenorhabditis elegans with two-photon fluorescence lifetime imaging. Front Chem 2023; 11:1161775. [PMID: 37123874 PMCID: PMC10137682 DOI: 10.3389/fchem.2023.1161775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Fat metabolism is an important modifier of aging and longevity in Caenorhabditis elegans. Given the anatomy and hermaphroditic nature of C. elegans, a major challenge is to distinguish fats that serve the energetic needs of the parent from those that are allocated to the progeny. Broadband coherent anti-Stokes Raman scattering (BCARS) microscopy has revealed that the composition and dynamics of lipid particles are heterogeneous both within and between different tissues of this organism. Using BCARS, we have previously succeeded in distinguishing lipid-rich particles that serve as energetic reservoirs of the parent from those that are destined for the progeny. While BCARS microscopy produces high-resolution images with very high information content, it is not yet a widely available platform. Here we report a new approach combining the lipophilic vital dye Nile Red and two-photon fluorescence lifetime imaging microscopy (2p-FLIM) for the in vivo discrimination of lipid particle sub-types. While it is widely accepted that Nile Red staining yields unreliable results for detecting lipid structures in live C. elegans due to strong interference of autofluorescence and non-specific staining signals, our results show that simple FLIM phasor analysis can effectively separate those signals and is capable of differentiating the non-polar lipid-dominant (lipid-storage), polar lipid-dominant (yolk lipoprotein) particles, and the intermediates that have been observed using BCARS microscopy. An advantage of this approach is that images can be acquired using common, commercially available 2p-FLIM systems within about 10% of the time required to generate a BCARS image. Our work provides a novel, broadly accessible approach for analyzing lipid-containing structures in a complex, live whole organism context.
Collapse
Affiliation(s)
- Wei-Wen Chen
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| | - Wenyu Tang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| | - Emily K. Hamerton
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| | - Penelope X. Kuo
- School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, United States
| | - George A. Lemieux
- School of Medicine, University of California, San Francisco, CA, United States
| | - Kaveh Ashrafi
- School of Medicine, University of California, San Francisco, CA, United States
| | - Marcus T. Cicerone
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
115
|
Vogt MC, Hobert O. Starvation-induced changes in somatic insulin/IGF-1R signaling drive metabolic programming across generations. SCIENCE ADVANCES 2023; 9:eade1817. [PMID: 37027477 PMCID: PMC10081852 DOI: 10.1126/sciadv.ade1817] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 03/08/2023] [Indexed: 05/30/2023]
Abstract
Exposure to adverse nutritional and metabolic environments during critical periods of development can exert long-lasting effects on health outcomes of an individual and its descendants. Although such metabolic programming has been observed in multiple species and in response to distinct nutritional stressors, conclusive insights into signaling pathways and mechanisms responsible for initiating, mediating, and manifesting changes to metabolism and behavior across generations remain scarce. By using a starvation paradigm in Caenorhabditis elegans, we show that starvation-induced changes in dauer formation-16/forkhead box transcription factor class O (DAF-16/FoxO) activity, the main downstream target of insulin/insulin-like growth factor 1 (IGF-1) receptor signaling, are responsible for metabolic programming phenotypes. Tissue-specific depletion of DAF-16/FoxO during distinct developmental time points demonstrates that DAF-16/FoxO acts in somatic tissues, but not directly in the germline, to both initiate and manifest metabolic programming. In conclusion, our study deciphers multifaceted and critical roles of highly conserved insulin/IGF-1 receptor signaling in determining health outcomes and behavior across generations.
Collapse
|
116
|
Yu Y, Chen J, An L, Huang T, Wang W, Cheng Z, Wang L, Xu X, Zhao Z, Fu X, Ma J. Knockdown of phosphatases of regenerating liver-1 prolongs the lifespan of Caenorhabditis elegans via activating DAF-16/FOXO. FASEB J 2023; 37:e22844. [PMID: 36906287 DOI: 10.1096/fj.202202003r] [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: 11/29/2022] [Revised: 01/20/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023]
Abstract
Phosphatases of regenerating liver (PRLs) are dual-specificity protein phosphatases. The aberrant expression of PRLs threatens human health, but their biological functions and pathogenic mechanisms are unclear yet. Herein, the structure and biological functions of PRLs were investigated using the Caenorhabditis elegans (C. elegans). Structurally, this phosphatase in C. elegans, named PRL-1, consisted of a conserved signature sequence WPD loop and a single C(X)5 R domain. Besides, by Western blot, immunohistochemistry and immunofluorescence staining, PRL-1 was proved to mainly express in larval stages and express in intestinal tissues. Afterward, by feeding-based RNA-interference method, knockdown of prl-1 prolonged the lifespan of C. elegans but also improved their healthspan, such as locomotion, pharyngeal pumping frequency, and defecation interval time. Furthermore, the above effects of prl-1 appeared to be taken without acting on germline signaling, diet restriction pathway, insulin/insulin-like growth factor 1 signaling pathway, and SIR-2.1 but through a DAF-16-dependent pathway. Moreover, knockdown of prl-1 induced the nuclear translocation of DAF-16, and upregulated the expression of daf-16, sod-3, mtl-1, and ctl-2. Finally, suppression of prl-1 also reduced the ROS. In conclusion, suppression of prl-1 enhanced the lifespan and survival quality of C. elegans, which provides a theoretical basis for the pathogenesis of PRLs in related human diseases.
Collapse
Affiliation(s)
- Yaoru Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jing Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lu An
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tianci Huang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Wenbo Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Ziqi Cheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xuesong Xu
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zhizhuang Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xueqi Fu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Junfeng Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
117
|
Gao SM, Qi Y, Zhang Q, Mohammed AS, Lee YT, Guan Y, Li H, Fu Y, Wang MC. Aging Atlas Reveals Cell-Type-Specific Regulation of Pro-longevity Strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530490. [PMID: 36909655 PMCID: PMC10002668 DOI: 10.1101/2023.02.28.530490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Organism aging occurs at the multicellular level; however, how pro-longevity mechanisms slow down aging in different cell types remains unclear. We generated single-cell transcriptomic atlases across the lifespan of Caenorhabditis elegans under different pro-longevity conditions (http://mengwanglab.org/atlas). We found cell-specific, age-related changes across somatic and germ cell types and developed transcriptomic aging clocks for different tissues. These clocks enabled us to determine tissue-specific aging-slowing effects of different pro-longevity mechanisms, and identify major cell types sensitive to these regulations. Additionally, we provided a systemic view of alternative polyadenylation events in different cell types, as well as their cell-type-specific changes during aging and under different pro-longevity conditions. Together, this study provides molecular insights into how aging occurs in different cell types and how they respond to pro-longevity strategies.
Collapse
|
118
|
Xiao Y, Liu F, Zhu X, Li S, Meng L, Jiang N, Yu C, Wang H, Qin Y, Hui J, Yu C, Liu Y. Dioscin integrates regulation of monosaturated fatty acid metabolism to extend the life span through XBP-1/SBP-1 dependent manner. iScience 2023; 26:106265. [PMID: 36936783 PMCID: PMC10014289 DOI: 10.1016/j.isci.2023.106265] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 02/17/2023] [Indexed: 03/11/2023] Open
Abstract
Delay aging, especially in healthy life extension, brought the most interest to the medical field. Searching for anti-aging drugs with relative safety profiles bring natural products in hotspot. In this study, we find that dioscin promotes the health span extension in wild-type Caenorhabditis elegans. Through the genetic screening in C. elegans, we further reveal that dioscin activates the transcription factor SBP-1/SREBP by the UPRER transcription factor XBP-1 to upregulate transcription of the Δ9 desaturase FAT-5 and FAT-7, resulting in increased monounsaturated fatty acid content which requires for healthy life span extension. Intriguingly, through tissue-specific knockdown, we find that dioscin modulates the health span by activating SBP-1 in the intestine. Unexpectedly, dietary supplementation of POA and OA rescues XBP-1, SBP-1 mutants-induced shortened life span phenotype. Considering the conservation of MUFAs metabolism, dioscin may promote health span in other species, including mammals. Our work suggests that dioscin might be a promising candidate for developing anti-aging agent.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Corresponding author
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xinting Zhu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Lingjie Meng
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Nian Jiang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Haijuan Wang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ying Qin
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jing Hui
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chunbo Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yun Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Corresponding author
| |
Collapse
|
119
|
Hernández-Cruz E, Eugenio-Pérez D, Ramírez-Magaña KJ, Pedraza-Chaverri J. Effects of Vegetal Extracts and Metabolites against Oxidative Stress and Associated Diseases: Studies in Caenorhabditis elegans. ACS OMEGA 2023; 8:8936-8959. [PMID: 36936291 PMCID: PMC10018526 DOI: 10.1021/acsomega.2c07025] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Oxidative stress is a natural physiological process where the levels of oxidants, such as reactive oxygen species (ROS) and nitrogen (RNS), exceed the strategy of antioxidant defenses, culminating in the interruption of redox signaling and control. Oxidative stress is associated with multiple pathologies, including premature aging, neurodegenerative diseases, obesity, diabetes, atherosclerosis, and arthritis. It is not yet clear whether oxidative stress is the cause or consequence of these diseases; however, it has been shown that using compounds with antioxidant properties, particularly compounds of natural origin, could prevent or slow down the progress of different pathologies. Within this context, the Caenorhabditis elegans (C. elegans) model has served to study the effect of different metabolites and natural compounds, which has helped to decipher molecular targets and the effect of these compounds on premature aging and some diseases such as neurodegenerative diseases and dyslipidemia. This article lists the studies carried out on C. elegans in which metabolites and natural extracts have been tested against oxidative stress and the pathologies associated with providing an overview of the discoveries in the redox area made with this nematode.
Collapse
Affiliation(s)
- Estefani
Yaquelin Hernández-Cruz
- Department
of Biology, Faculty of Chemistry, National
Autonomous University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
- Postgraduate
in Biological Sciences, National Autonomous
University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Dianelena Eugenio-Pérez
- Department
of Biology, Faculty of Chemistry, National
Autonomous University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
- Postgraduate
in Biochemical Sciences, National Autonomous
University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Karla Jaqueline Ramírez-Magaña
- Department
of Biology, Faculty of Chemistry, National
Autonomous University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
- Postgraduate
in Biochemical Sciences, National Autonomous
University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - José Pedraza-Chaverri
- Department
of Biology, Faculty of Chemistry, National
Autonomous University of Mexico, Ciudad Universitaria, 04510 Mexico City, Mexico
| |
Collapse
|
120
|
Makhijani P, Basso PJ, Chan YT, Chen N, Baechle J, Khan S, Furman D, Tsai S, Winer DA. Regulation of the immune system by the insulin receptor in health and disease. Front Endocrinol (Lausanne) 2023; 14:1128622. [PMID: 36992811 PMCID: PMC10040865 DOI: 10.3389/fendo.2023.1128622] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
The signaling pathways downstream of the insulin receptor (InsR) are some of the most evolutionarily conserved pathways that regulate organism longevity and metabolism. InsR signaling is well characterized in metabolic tissues, such as liver, muscle, and fat, actively orchestrating cellular processes, including growth, survival, and nutrient metabolism. However, cells of the immune system also express the InsR and downstream signaling machinery, and there is increasing appreciation for the involvement of InsR signaling in shaping the immune response. Here, we summarize current understanding of InsR signaling pathways in different immune cell subsets and their impact on cellular metabolism, differentiation, and effector versus regulatory function. We also discuss mechanistic links between altered InsR signaling and immune dysfunction in various disease settings and conditions, with a focus on age related conditions, such as type 2 diabetes, cancer and infection vulnerability.
Collapse
Affiliation(s)
- Priya Makhijani
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
| | - Paulo José Basso
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yi Tao Chan
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nan Chen
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jordan Baechle
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Saad Khan
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - David Furman
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1, 000 Immunomes Project, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pilar, Argentina
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel A. Winer
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
121
|
Wang Y, Arnold ML, Smart AJ, Wang G, Androwski RJ, Morera A, Nguyen KCQ, Schweinsberg PJ, Bai G, Cooper J, Hall DH, Driscoll M, Grant BD. Large vesicle extrusions from C. elegans neurons are consumed and stimulated by glial-like phagocytosis activity of the neighboring cell. eLife 2023; 12:e82227. [PMID: 36861960 PMCID: PMC10023159 DOI: 10.7554/elife.82227] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/28/2023] [Indexed: 03/03/2023] Open
Abstract
Caenorhabditis elegans neurons under stress can produce giant vesicles, several microns in diameter, called exophers. Current models suggest that exophers are neuroprotective, providing a mechanism for stressed neurons to eject toxic protein aggregates and organelles. However, little is known of the fate of the exopher once it leaves the neuron. We found that exophers produced by mechanosensory neurons in C. elegans are engulfed by surrounding hypodermal skin cells and are then broken up into numerous smaller vesicles that acquire hypodermal phagosome maturation markers, with vesicular contents gradually degraded by hypodermal lysosomes. Consistent with the hypodermis acting as an exopher phagocyte, we found that exopher removal requires hypodermal actin and Arp2/3, and the hypodermal plasma membrane adjacent to newly formed exophers accumulates dynamic F-actin during budding. Efficient fission of engulfed exopher-phagosomes to produce smaller vesicles and degrade their contents requires phagosome maturation factors SAND-1/Mon1, GTPase RAB-35, the CNT-1 ARF-GAP, and microtubule motor-associated GTPase ARL-8, suggesting a close coupling of phagosome fission and phagosome maturation. Lysosome activity was required to degrade exopher contents in the hypodermis but not for exopher-phagosome resolution into smaller vesicles. Importantly, we found that GTPase ARF-6 and effector SEC-10/exocyst activity in the hypodermis, along with the CED-1 phagocytic receptor, is required for efficient production of exophers by the neuron. Our results indicate that the neuron requires specific interaction with the phagocyte for an efficient exopher response, a mechanistic feature potentially conserved with mammalian exophergenesis, and similar to neuronal pruning by phagocytic glia that influences neurodegenerative disease.
Collapse
Affiliation(s)
- Yu Wang
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Anna Joelle Smart
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Guoqiang Wang
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Rebecca J Androwski
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Andres Morera
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Ken CQ Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, BronxNew YorkUnited States
| | - Peter J Schweinsberg
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Ge Bai
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Jason Cooper
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, BronxNew YorkUnited States
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers UniversityPiscatawayUnited States
- Rutgers Center for Lipid ResearchNew BrunswickUnited States
| |
Collapse
|
122
|
Wang J, Zhou Y, Yu Y, Wang Y, Xue D, Zhou Y, Li X. A ginseng-derived rhamnogalacturonan I (RG-I) pectin promotes longevity via TOR signalling in Caenorhabditis elegans. Carbohydr Polym 2023; 312:120818. [PMID: 37059546 DOI: 10.1016/j.carbpol.2023.120818] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Panax ginseng C. A. Meyer (ginseng), a traditional Chinese herb, is usually used to improve health and increase anti-aging activity for human. Polysaccharides are bioactive components of ginseng. Herein, using Caenorhabditis elegans as a model, we discovered a ginseng-derived rhamnogalacturonan I (RG-I) pectin WGPA-1-RG promoted longevity via TOR signalling pathway with transcription factors FOXO/DAF-16 and Nrf2/SKN-1 accumulated in the nucleus, where they activated target genes. And the WGPA-1-RG-mediated lifespan extension was dependent on endocytosis, rather than a bacterial metabolic process. Glycosidic linkage analyses combined with arabinose- and galactose-releasing enzyme hydrolyses identified the RG-I backbone of WGPA-1-RG was primarily substituted with α-1,5-linked arabinan, β-1,4-linked galactan and arabinogalactan II (AG-II) side chains. Feeding worms with the WGPA-1-RG-derived fractions which lost distinct structural elements by enzymatic digestions, we found the arabinan side chains prominently contributed to the longevity-promoting activity of WGPA-1-RG. These findings provide a novel ginseng-derived nutrient that potentially increases human longevity.
Collapse
|
123
|
Ma J, Liu Z, Gao X, Bao Y, Hong Y, He X, Zhu W, Li Y, Huang W, Zheng N, Sheng L, Zhou B, Chen H, Li H. Gut microbiota remodeling improves natural aging-related disorders through Akkermansia muciniphila and its derived acetic acid. Pharmacol Res 2023; 189:106687. [PMID: 36746362 DOI: 10.1016/j.phrs.2023.106687] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/05/2023]
Abstract
Accumulating evidence indicates gut microbiota contributes to aging-related disorders. However, the exact mechanism underlying gut dysbiosis-related pathophysiological changes during aging remains largely unclear. In the current study, we first performed gut microbiota remodeling on old mice by fecal microbiota transplantation (FMT) from young mice, and then characterized the bacteria signature that was specifically altered by FMT. Our results revealed that FMT significantly improved natural aging-related systemic disorders, particularly exerted hepatoprotective effects, and improved glucose sensitivity, hepatosplenomegaly, inflammaging, antioxidative capacity and intestinal barrier. Moreover, FMT particularly increased the abundance of fecal A.muciniphila, which was almost nondetectable in old mice. Interestingly, A.muciniphila supplementation also exerted similar benefits with FMT on old mice. Notably, targeted metabolomics on short chain fatty acids (SCFAs) revealed that only acetic acid was consistently reversed by FMT. Then, acetic acid intervention exerted beneficial actions on both Caenorhabditis elegans and natural aging mice. In conclusion, our current study demonstrated that gut microbiota remodeling improved natural aging-related disorders through A.muciniphila and its derived acetic acid, suggesting that interventions with potent stimulative capacity on A. muciniphila growth and production of acetic acid was alternative and effective way to maintain healthy aging. DATA AVAILABILITY STATEMENT: The data of RNAseq and 16 S rRNA gene sequencing can be accessed in NCBI with the accession number PRJNA848996 and PRJNA849355.
Collapse
Affiliation(s)
- Junli Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zekun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinxin Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyang Bao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying Hong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaofang He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ben Zhou
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, 200025, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
124
|
Wang H, Brey CW, Wang Y, Gaugler R, Hashmi S. KLF regulation of insulin pathway genes. 3 Biotech 2023; 13:87. [PMID: 36816753 PMCID: PMC9935763 DOI: 10.1007/s13205-023-03502-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
Alteration in lipid metabolism can result in fat accumulation in adipose tissues, which may lead to two most important human diseases, obesity and diabetes. A shift in lipid metabolism deregulates signaling pathways which regulates obesity and/or diabetes. In this study, we examined the components of insulin/ TGF-β pathways and their genetic interaction with Krüppel-like transcription factors (KLFs). Their role in energy homeostasis were discussed. We separately created klf/daf genes double mutants by carrying out klfs RNAi on daf-2 (e1391), daf-4 (e1364), daf-7 (e1372); dpy-1 (e1), daf-14 (m77), daf-16 (mgDf50) mutants. And then conducted Oil O Red staining to assay the klf/daf RNAi worms for fat deposits and examine genetic interaction between klfs and daf genes. The results showed that worms bearing klf-1, 2, or 3 and daf-2, or daf-4 mutations deposit large, but similar fat levels as individual mutants. The results suggested that they target the same molecular pathway of fat storage. klf-1, 2 or 3 RNAi /daf-7 worms showed higher fat deposits in klf-1, 2, or 3 RNAi/daf-7 worms than klf-1, 2, or 3 RNAi or daf-7 mutants alone, which showed a functional interaction between klfs and daf-7 in perhaps TGF-β-like pathway. Altogether our study suggests a direct role of klfs in insulin signaling pathway.
Collapse
Affiliation(s)
- Huan Wang
- College of Bioscience and Biotechnology, Shenyang Agriculture University, Shanyang, 110866 China
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, USA
| | | | - Yi Wang
- Laboratory of Developmental Biology, Center for Vector Biology, Rutgers University, 180 Jones Avenue, New Brunswick, NJ 08901 USA
| | - Randy Gaugler
- Laboratory of Developmental Biology, Center for Vector Biology, Rutgers University, 180 Jones Avenue, New Brunswick, NJ 08901 USA
| | - Sarwar Hashmi
- Laboratory of Developmental Biology, Center for Vector Biology, Rutgers University, 180 Jones Avenue, New Brunswick, NJ 08901 USA
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, USA
| |
Collapse
|
125
|
Moldakozhayev A, Gladyshev VN. Metabolism, homeostasis, and aging. Trends Endocrinol Metab 2023; 34:158-169. [PMID: 36681595 PMCID: PMC11096277 DOI: 10.1016/j.tem.2023.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/21/2023]
Abstract
We propose a two-mode (pursuit/maintenance) model of metabolism defined by usable resource availability. Pursuit, consisting of anabolism and catabolism, dominates when usable resources are plentiful and leads to the generation of metabolic waste. In turn, maintenance of a system is activated by elevated metabolic waste during resource depletion. Interaction with the environment results in pendulum-like swings between these metabolic states in thriveless attempts to maintain the least deleterious organismal state - ephemeral homeostasis. Imperfectness of biological processes during these attempts supports the accumulation of the deleteriome, driving organismal aging. We discuss how metabolic adjustment by the environment and resource stabilization may modulate healthspan and lifespan.
Collapse
Affiliation(s)
- Alibek Moldakozhayev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, QC H3A 2B4, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, QC H4A 3J1, Canada
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
126
|
Brandel-Ankrapp KL, Arey RN. Uncovering novel regulators of memory using C. elegans genetic and genomic analysis. Biochem Soc Trans 2023; 51:161-171. [PMID: 36744642 PMCID: PMC10518207 DOI: 10.1042/bst20220455] [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: 10/14/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
How organisms learn and encode memory is an outstanding question in neuroscience research. Specifically, how memories are acquired and consolidated at the level of molecular and gene pathways remains unclear. In addition, memory is disrupted in a wide variety of neurological disorders; therefore, discovering molecular regulators of memory may reveal therapeutic targets for these disorders. C. elegans are an excellent model to uncover molecular and genetic regulators of memory. Indeed, the nematode's invariant neuronal lineage, fully mapped genome, and conserved associative behaviors have allowed the development of a breadth of genetic and genomic tools to examine learning and memory. In this mini-review, we discuss novel and exciting genetic and genomic techniques used to examine molecular and genetic underpinnings of memory from the level of the whole-worm to tissue-specific and cell-type specific approaches with high spatiotemporal resolution.
Collapse
Affiliation(s)
- Katie L. Brandel-Ankrapp
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
| | - Rachel N. Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
127
|
Xiao Y, Zhang L, Zhu X, Qin Y, Yu C, Jiang N, Li S, Liu F, Liu Y. Luteolin promotes pathogen resistance in Caenorhabditis elegans via DAF-2/DAF-16 insulin-like signaling pathway. Int Immunopharmacol 2023; 115:109679. [PMID: 36640711 DOI: 10.1016/j.intimp.2023.109679] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/20/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023]
Abstract
The DAF-2/DAF-16 insulin-like signaling pathway was an evolutionarily conserved pathway, which regulated many aspects of organismal physiology, such as pathogen resistance, metabolism, stress response, longevity. Luteolin, a flavone contained in many medical plants and in vegetables, had been shown to exhibit activities such as anti-tumor, anti-oxidant and neuroprotective effects. However, whether the Luteolin influenced the immune response and the underlying molecular mechanisms remained obscure. We found that Luteolin increased resistance to not only the Gram-negative pathogens Pseudomonas aeruginosa and Salmonella enterica but also the Gram-positive pathogens Enterococcus faecalis and Staphylococcus aureus in dose dependent manner. Meanwhile, Luteolin promoted host immune response via inhibiting the growth of pathogenic bacteria. Through the genetic screening in C. elegans, we found that Luteolin promoted innate immunity via DAF-2/DAF-16 insulin-like signaling pathway rather than p38 MAPK pathway and SKN-1. Furthermore, Luteolin activated the DAF-16/FOXO transcription factor for innate immune response. Our work suggested that Luteolin had the potential of improving the patients with pathogen infection.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Li Zhang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xinting Zhu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ying Qin
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Nian Jiang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Yun Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Center of Forensic Expertise, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
128
|
He Y, Hao F, Fu H, Tian G, Zhang Y, Fu K, Qi B. N-glycosylated intestinal protein BCF-1 shapes microbial colonization by binding bacteria via its fimbrial protein. Cell Rep 2023; 42:111993. [PMID: 36662624 DOI: 10.1016/j.celrep.2023.111993] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/06/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
Microbial colonization plays an instrumental role in the health of the host. However, the host factors that facilitate the establishment of the microbial colonization remain unclear. Here, we establish a screening method to identify host factors regulating E. coli colonization in C. elegans. We find that a BCF-1 possessing N-glycosylation promotes E. coli colonization by directly binding to E. coli via its fimbrial protein, YdeR. BCF-1 is activated by the bacteria and interacts with an oligosaccharyl transferase, OSTB-1, which is critical for regulating E. coli colonization. We also show that the N-glycosylation of BCF-1 is critical for E. coli colonization. In addition, we find that the microbiota composition is shaped by BCF-1. In summary, this study shows a "scaffold model" for bacterial colonization between a host glycoprotein and E. coli, and it also introduces a powerful research approach to identify individual host factors involved in modulating bacterial colonization.
Collapse
Affiliation(s)
- Yongjuan He
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Fanrui Hao
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Herui Fu
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Guojing Tian
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Yingyang Zhang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Kai Fu
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Bin Qi
- Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China.
| |
Collapse
|
129
|
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
|
130
|
Tang LTH, Lee GA, Cook SJ, Ho J, Potter CC, Bülow HE. Restructuring of an asymmetric neural circuit during associative learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523604. [PMID: 36711870 PMCID: PMC9882173 DOI: 10.1101/2023.01.12.523604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Asymmetric brain function is common across the animal kingdom and involved in language processing, and likely in learning and memory. What regulates asymmetric brain function remains elusive. Here, we show that the nematode Caenorhabditis elegans restructures an asymmetric salt sensing neural circuit during associative learning. Worms memorize and prefer the salt concentration at which they were raised in the presence of food through a left-biased network architecture. When conditioned at elevated salt concentrations, animals change the left-biased to a right-biased network, which explains the changed salt-seeking behavior. The changes in circuit architecture require new synapse formation induced through asymmetric, paracrine insulin-signaling. Therefore, experience-dependent changes in asymmetric network architecture rely on paracrine insulin signaling and are fundamental to learning and behavior.
Collapse
|
131
|
Application of Caenorhabditis elegans in Lipid Metabolism Research. Int J Mol Sci 2023; 24:ijms24021173. [PMID: 36674689 PMCID: PMC9860639 DOI: 10.3390/ijms24021173] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Over the last decade, the development and prevalence of obesity have posed a serious public health risk, which has prompted studies on the regulation of adiposity. With the ease of genetic manipulation, the diversity of the methods for characterizing body fat levels, and the observability of feeding behavior, Caenorhabditis elegans (C. elegans) is considered an excellent model for exploring energy homeostasis and the regulation of the cellular fat storage. In addition, the homology with mammals in the genes related to the lipid metabolism allows many aspects of lipid modulation by the regulators of the central nervous system to be conserved in this ideal model organism. In recent years, as the complex network of genes that maintain an energy balance has been gradually expanded and refined, the regulatory mechanisms of lipid storage have become clearer. Furthermore, the development of methods and devices to assess the lipid levels has become a powerful tool for studies in lipid droplet biology and the regulation of the nematode lipid metabolism. Herein, based on the rapid progress of C. elegans lipid metabolism-related studies, this review outlined the lipid metabolic processes, the major signaling pathways of fat storage regulation, and the primary experimental methods to assess the lipid content in nematodes. Therefore, this model system holds great promise for facilitating the understanding, management, and therapies of human obesity and other metabolism-related diseases.
Collapse
|
132
|
Estrada-Valencia R, de Lima ME, Colonnello A, Rangel-López E, Saraiva NR, de Ávila DS, Aschner M, Santamaría A. The Endocannabinoid System in Caenorhabditis elegans. Rev Physiol Biochem Pharmacol 2023; 184:1-31. [PMID: 34401955 PMCID: PMC8850531 DOI: 10.1007/112_2021_64] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The existence of a formal Endocannabinoid System in C. elegans has been questioned due to data showing the absence of typical cannabinoid receptors in the worm; however, the presence of a full metabolism for endocannabinoids, alternative ligands, and receptors for these agents and a considerable number of orthologous and homologous genes regulating physiological cannabinoid-like signals and responses - several of which are similar to those of mammals - demonstrates a well-structured and functional complex system in nematodes. In this review, we describe and compare similarities and differences between the Endocannabinoid System in mammals and nematodes, highlighting the basis for the integral study of this novel system in the worm.
Collapse
Affiliation(s)
| | - María Eduarda de Lima
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Aline Colonnello
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Edgar Rangel-López
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Nariani Rocha Saraiva
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Daiana Silva de Ávila
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico.
| |
Collapse
|
133
|
Fidalgo S, Yeoman MS. Age-Related Changes in Central Nervous System 5-Hydroxytryptamine Signalling and Its Potential Effects on the Regulation of Lifespan. Subcell Biochem 2023; 102:379-413. [PMID: 36600141 DOI: 10.1007/978-3-031-21410-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Serotonin or 5-hydroxytryptamine (5-HT) is an important neurotransmitter in the central nervous system and the periphery. Most 5-HT (~99%) is found in the periphery where it regulates the function of the gastrointestinal (GI) tract and is an important regulator of platelet aggregation. However, the remaining 1% that is found in the central nervous system (CNS) can regulate a range of physiological processes such as learning and memory formation, mood, food intake, sleep, temperature and pain perception. More recent work on the CNS of invertebrate model systems has shown that 5-HT can directly regulate lifespan.This chapter will focus on detailing how CNS 5-HT signalling is altered with increasing age and the potential consequences this has on its ability to regulate lifespan.
Collapse
Affiliation(s)
| | - Mark S Yeoman
- Centre for Stress and Age-Related Disease, School of Applied Sciences, University of Brighton, Brighton, United Kingdom.
| |
Collapse
|
134
|
Kunizheva SS, Volobaev VP, Plotnikova MY, Kupriyanova DA, Kuznetsova IL, Tyazhelova TV, Rogaev EI. Current Trends and Approaches to the Search for Genetic Determinants of Aging and Longevity. RUSS J GENET+ 2022; 58:1427-1443. [PMID: 36590179 PMCID: PMC9794410 DOI: 10.1134/s1022795422120067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/29/2022]
Abstract
Aging is a natural process of extinction of the body and the main aspect that determines the life expectancy for individuals who have survived to the post-reproductive period. The process of aging is accompanied by certain physiological, immune, and metabolic changes in the body, as well as the development of age-related diseases. The contribution of genetic factors to human life expectancy is estimated at about 25-30%. Despite the success in identifying genes and metabolic pathways that may be involved in the life extension process in model organisms, the key question remains to what extent these data can be extrapolated to humans, for example, because of the complexity of its biological and sociocultural systems, as well as possible species differences in life expectancy and causes of mortality. New molecular genetic methods have significantly expanded the possibilities for searching for genetic factors of human life expectancy and identifying metabolic pathways of aging, the interaction of genes and transcription factors, the regulation of gene expression at the level of transcription, and epigenetic modifications. The review presents the latest research and current strategies for studying the genetic basis of human aging and longevity: the study of individual candidate genes in genetic population studies, variations identified by the GWAS method, immunogenetic differences in aging, and genomic studies to identify factors of "healthy aging." Understanding the mechanisms of the interaction between factors affecting the life expectancy and the possibility of their regulation can become the basis for developing comprehensive measures to achieve healthy longevity. Supplementary Information The online version contains supplementary material available at 10.1134/S1022795422120067.
Collapse
Affiliation(s)
- S. S. Kunizheva
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - V. P. Volobaev
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - M. Yu. Plotnikova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
| | - D. A. Kupriyanova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - I. L. Kuznetsova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - T. V. Tyazhelova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - E. I. Rogaev
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- University of Massachusetts Chan Medical School, 01545 Shrewsbury, MA United States
| |
Collapse
|
135
|
Planococcus maritimu ML1206 Strain Enhances Stress Resistance and Extends the Lifespan in Caenorhabditis elegans via FOXO/DAF-16. Mar Drugs 2022; 21:md21010001. [PMID: 36662174 PMCID: PMC9866299 DOI: 10.3390/md21010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The antioxidant effect of probiotics has been widely recognized across the world, which is of great significance in food, medicine, and aquaculture. There are abundant marine microbial resources in the ocean, which provide a new space for humans to explore new probiotics. Previously, we reported on the anti-infective effects of Planococcus maritimu ML1206, a potential marine probiotic. The antioxidant activity of ML1206 in C. elegans was studied in this paper. The study showed that ML1206 could improve the ability of nematodes to resist oxidative stress and effectively prolong their lifespan. The results confirmed that ML1206 could significantly increase the activities of CAT and GSH-PX, and reduce the accumulation of reactive oxygen species (ROS) in nematodes under oxidative stress conditions. In addition, ML1206 promoted DAF-16 transfer to the nucleus and upregulated the expression of sod-3, hsp-16.2, and ctl-2, which are downstream antioxidant-related genes of DAF-16. Furthermore, the expression of the SOD-3::GFP and HSP-16.2::GFP was significantly higher in the transgenic strains fed with ML1206 than that in the control group fed with OP50, with or without stress. In summary, these findings suggest that ML1206 is a novel marine probiotic with an antioxidant function that stimulates nematodes to improve their defense abilities against oxidative stress and prolong the lifespan by regulating the translocation of FOXO/DAF-16. Therefore, ML1206 may be explored as a potential dietary supplement in aquaculture and for anti-aging and antioxidant purposes.
Collapse
|
136
|
Mitochondrial function and nutrient sensing pathways in ageing: enhancing longevity through dietary interventions. Biogerontology 2022; 23:657-680. [PMID: 35842501 DOI: 10.1007/s10522-022-09978-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
Ageing is accompanied by alterations in several biochemical processes, highly influenced by its environment. It is controlled by the interactions at various levels of biological hierarchy. To maintain homeostasis, a number of nutrient sensors respond to the nutritional status of the cell and control its energy metabolism. Mitochondrial physiology is influenced by the energy status of the cell. The alterations in mitochondrial physiology and the network of nutrient sensors result in mitochondrial damage leading to age related metabolic degeneration and diseases. Calorie restriction (CR) has proved to be as the most successful intervention to achieve the goal of longevity and healthspan. CR elicits a hormetic response and regulates metabolism by modulating these networks. In this review, the authors summarize the interdependent relationship between mitochondrial physiology and nutrient sensors during the ageing process and their role in regulating metabolism.
Collapse
|
137
|
Yang W, Xia W, Zheng B, Li T, Liu RH. DAF-16 is involved in colonic metabolites of ferulic acid-promoted longevity and stress resistance of Caenorhabditis elegans. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:7017-7029. [PMID: 35689482 DOI: 10.1002/jsfa.12063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/11/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Ferulic acid (FA) is a dietary polyphenol widely found in plant tissues. It has long been considered to have health-promoting qualities. However, the biological properties of dietary polyphenols depend largely on their absorption during digestion, and the effects of their intestinal metabolites on human health have attracted the interest of researchers. This study evaluated the effects of three main colonic metabolites of FA - 3-(3,4-dihydroxyphenyl)propionic acid (3,4diOHPPA), 3-(3-hydroxyphenyl)propionic acid (3OHPPA) and 3-phenylpropionic acid (3PPA) - on longevity and stress resistance in Caenorhabditis elegans. RESULTS Our results showed that 3,4diOHPPA, 3OHPPA and 3PPA extended the lifespan under normal conditions in C. elegans whereas FA did not. High doses of 3,4diOHPPA (0.5 mmol L-1 ), 3OHPPA (2.5 mmol L-1 ) and 3PPA (2.5 mmol L-1 ) prolonged the mean lifespan by 11.2%, 13.0% and 10.6%, respectively. Moreover, 3,4diOHPPA, 3OHPPA and 3PPA treatments promoted stress tolerance against heat, UV irradiation and paraquat. Furthermore, three metabolites ameliorated physical functions, including reactive oxygen species and malondialdehyde levels, motility and pharyngeal pumping rate. The anti-aging activities mediated by 3,4diOHPPA, 3OHPPA and 3PPA depend on the HSF-1 and JNK-1 linked insulin/IGF-1 signaling pathway, which converge onto DAF-16. CONCLUSION The current findings suggest that colonic metabolites of FA have the potential for use as anti-aging bioactivate compounds. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenhan Yang
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Wen Xia
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Bisheng Zheng
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong ERA Food and Life Health Research Institute, Guangzhou, China
| | - Tong Li
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Rui Hai Liu
- Department of Food Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
138
|
Comparing dormancy in two distantly related tunicates reveals morphological, molecular, and ecological convergences and repeated co-option. Sci Rep 2022; 12:12620. [PMID: 35871255 PMCID: PMC9308810 DOI: 10.1038/s41598-022-16656-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/13/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractMany asexually-propagating marine invertebrates can survive extreme environmental conditions by developing dormant structures, i.e., morphologically simplified bodies that retain the capacity to completely regenerate a functional adult when conditions return to normal. Here, we examine the environmental, morphological, and molecular characteristics of dormancy in two distantly related clonal tunicate species: Polyandrocarpa zorritensis and Clavelina lepadiformis. In both species, we report that the dormant structures are able to withstand harsher temperature and salinity conditions compared to the adults. The dormant structures are the dominant forms these species employ to survive adverse conditions when the zooids themselves cannot survive. While previous work shows C. lepadiformis dormant stage is present in winters in the Atlantic Ocean and summers in the Mediterranean, this study is the first to show a year-round presence of P. zorritensis dormant forms in NW Italy, even in the late winter when all zooids have disappeared. By finely controlling the entry and exit of dormancy in laboratory-reared individuals, we were able to select and characterize the morphology of dormant structures associated with their transcriptome dynamics. In both species, we identified putative stem and nutritive cells in structures that resemble the earliest stages of asexual propagation. By characterizing gene expression during dormancy and regeneration into the adult body plan (i.e., germination), we observed that genes which control dormancy and environmental sensing in other metazoans, notably HIF-α and insulin signaling genes, are also expressed in tunicate dormancy. Germination-related genes in these two species, such as the retinoic acid pathway, are also found in other unrelated clonal tunicates during asexual development. These results are suggestive of repeated co-option of conserved eco-physiological and regeneration programs for the origin of novel dormancy-germination processes across distantly related animal taxa.
Collapse
|
139
|
Lee H, Lee SJV. Recent Progress in Regulation of Aging by Insulin/IGF-1 Signaling in Caenorhabditis elegans. Mol Cells 2022; 45:763-770. [PMID: 36380728 PMCID: PMC9676989 DOI: 10.14348/molcells.2022.0097] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/18/2022] Open
Abstract
Caenorhabditis elegans has been used as a major model organism to identify genetic factors that regulate organismal aging and longevity. Insulin/insulin-like growth factor 1 (IGF- 1) signaling (IIS) regulates aging in many species, ranging from nematodes to humans. C. elegans is a nonpathogenic genetic nematode model, which has been extensively utilized to identify molecular and cellular components that function in organismal aging and longevity. Here, we review the recent progress in the role of IIS in aging and longevity, which involves direct regulation of protein and RNA homeostasis, stress resistance, metabolism and the activities of the endocrine system. We also discuss recently identified genetic factors that interact with canonical IIS components to regulate aging and health span in C. elegans. We expect this review to provide valuable insights into understanding animal aging, which could eventually help develop anti-aging drugs for humans.
Collapse
Affiliation(s)
- Hanseul Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seung-Jae V. Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
140
|
Haynes CM, Hekimi S. Mitochondrial dysfunction, aging, and the mitochondrial unfolded protein response in Caenorhabditis elegans. Genetics 2022; 222:iyac160. [PMID: 36342845 PMCID: PMC9713405 DOI: 10.1093/genetics/iyac160] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
We review the findings that establish that perturbations of various aspects of mitochondrial function, including oxidative phosphorylation, can promote lifespan extension, with different types of perturbations acting sometimes independently and additively on extending lifespan. We also review the great variety of processes and mechanisms that together form the mitochondrial unfolded protein response. We then explore the relationships between different types of mitochondrial dysfunction-dependent lifespan extension and the mitochondrial unfolded protein response. We conclude that, although several ways that induce extended lifespan through mitochondrial dysfunction require a functional mitochondrial unfolded protein response, there is no clear indication that activation of the mitochondrial unfolded protein response is sufficient to extend lifespan, despite the fact that the mitochondrial unfolded protein response impacts almost every aspect of mitochondrial function. In fact, in some contexts, mitochondrial unfolded protein response activation is deleterious. To explain this pattern, we hypothesize that, although triggered by mitochondrial dysfunction, the lifespan extension observed might not be the result of a change in mitochondrial function.
Collapse
Affiliation(s)
- Cole M Haynes
- Molecular, Cell and Cancer Biology, UMass-Chan Medical School, Worcester, MA 01655, USA
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
141
|
Haroon, Li YX, Ye CX, Su J, Nabi G, Su XH, Xing LX. De Novo Transcriptome Assembly and Analysis of Longevity Genes Using Subterranean Termite ( Reticulitermes chinensis) Castes. Int J Mol Sci 2022; 23:13660. [PMID: 36362447 PMCID: PMC9657995 DOI: 10.3390/ijms232113660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
The longevity phenomenon is entirely controlled by the insulin signaling pathway (IIS-pathway). Both vertebrates and invertebrates have IIS-pathways that are comparable to one another, though no one has previously described de novo transcriptome assembly of IIS-pathway-associated genes in termites. In this research, we analyzed the transcriptomes of both reproductive (primary kings “PK” and queens “PQ”, secondary worker reproductive kings “SWRK” and queens “SWRQ”) and non-reproductive (male “WM” and female “WF” workers) castes of the subterranean termite Reticulitermes chinensis. The goal was to identify the genes responsible for longevity in the reproductive and non-reproductive castes. Through transcriptome analysis, we annotated 103,589,264 sequence reads and 184,436 (7G) unigenes were assembled, GC performance was measured at 43.02%, and 64,046 sequences were reported as CDs sequences. Of which 35 IIS-pathway-associated genes were identified, among 35 genes, we focused on the phosphoinositide-dependent kinase-1 (Pdk1), protein kinase B2 (akt2-a), tuberous sclerosis-2 (Tsc2), mammalian target of rapamycin (mTOR), eukaryotic translation initiation factor 4E (EIF4E) and ribosomal protein S6 (RPS6) genes. Previously these genes (Pdk1, akt2-a, mTOR, EIF4E, and RPS6) were investigated in various organisms, that regulate physiological effects, growth factors, protein translation, cell survival, proliferation, protein synthesis, cell metabolism and survival, autophagy, fecundity rate, egg size, and follicle number, although the critical reason for longevity is still unclear in the termite castes. However, based on transcriptome profiling, the IIS-pathway-associated genes could prolong the reproductive caste lifespan and health span. Therefore, the transcriptomic shreds of evidence related to IIS-pathway genes provide new insights into the maintenance and relationships between biomolecular homeostasis and remarkable longevity. Finally, we propose a strategy for future research to decrypt the hidden costs associated with termite aging in reproductive and non-reproductive castes.
Collapse
Affiliation(s)
- Haroon
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Yu-Xin Li
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Chen-Xu Ye
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Jian Su
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, 31120 Krakow, Poland
| | - Xiao-Hong Su
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China
| | - Lian-Xi Xing
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China
| |
Collapse
|
142
|
Luo X, Zhang Y, Lu C, Zhang J. Role of insulin signaling pathway in apoptosis induced by food chain delivery of nano-silver under the action of environmental factors. Comp Biochem Physiol C Toxicol Pharmacol 2022; 261:109429. [PMID: 35944823 DOI: 10.1016/j.cbpc.2022.109429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate how the environmental factor affects the delivery of nano silver through food chain, we set up a two-stage food delivery chain model of Escherichia coli and Caenorhabditis elegans system. METHODS Through a two-stage food delivery chain model of E. coli and C. elegans, the mRNA expression levels of DAF-2, age-1, PDK-1, Akt-1 and DAF-16 in the insulin growth factor 1 signaling pathway in nematode gonad cells which occurs AgNPs induced apoptosis were evaluated and the apoptosis of gonad cells in the mutant strains of the above key genes were detected. RESULTS DAF-2, age-1, PDK-1 and Akt-1 could significantly negatively regulate the apoptosis of nematode cells induced by AgNPs, while DAF-16 could significantly promote the apoptosis induced by AgNPs. The DAF-16 up-regulated expression was a protective effect on the body and the phenomenon of DNA double-strand breaks was significantly increased. The damage effect induced by AgNPs was significantly enhanced in the presence of the environmental factor fulvic acid. CONCLUSION The damage effect induced by AgNPs after food delivery involves the regulation of the insulin growth factor 1 signaling pathway and environmental factors have a significant impact on the biological effects.
Collapse
Affiliation(s)
- Xun Luo
- School of Biological Engineering, Huainan Normal University, China.
| | - Yajun Zhang
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Health and Safety, Ministry of Education, China; Medicine School, Anhui University of Science & Technology, China.
| | - Changjie Lu
- School of Biological Engineering, Huainan Normal University, China
| | - Jiaming Zhang
- School of Biological Engineering, Huainan Normal University, China
| |
Collapse
|
143
|
Cheng D, Lee JS, Brown M, Ebert MS, McGrath PT, Tomioka M, Iino Y, Bargmann CI. Insulin/IGF signaling regulates presynaptic glutamate release in aversive olfactory learning. Cell Rep 2022; 41:111685. [DOI: 10.1016/j.celrep.2022.111685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/06/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
|
144
|
Zhang J, Zhao Y, Sun Z, Sun T. Lacticaseibacillus rhamnosus Probio-M9 extends the lifespan of Caenorhabditis elegans. Commun Biol 2022; 5:1139. [PMID: 36302976 PMCID: PMC9613993 DOI: 10.1038/s42003-022-04031-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
Abstract
Probiotics have been characterized as useful for maintaining the balance of host gut flora and conferring health effects, but few studies have focused on their potential for delaying aging in the host. Here we show that Lacticaseibacillus rhamnosus Probio-M9 (Probio-M9), a healthy breast milk probiotic, enhances the locomotor ability and slows the decline in muscle function of the model organism Caenorhabditis elegans. Live Probio-M9 significantly extends the lifespan of C. elegans in a dietary restriction-independent manner. By screening various aging-related mutants of C. elegans, we find that Probio-M9 extends lifespan via p38 cascade and daf-2 signaling pathways, independent on daf-16 but dependent on skn-1. Probio-M9 protects and repairs damaged mitochondria by activating mitochondrial unfolded protein response. The significant increase of amino acids, sphingolipid, galactose and fatty acids in bacterial metabolites might be involved in extending the lifespan of C. elegans. We reveal that Probio-M9 as a dietary supplementation had the potential to delay aging in C. elegans and also provide new methods and insights for further analyzing probiotics in improving host health and delaying the occurrence of age-related chronic diseases.
Collapse
Affiliation(s)
- Juntao Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Yanmei Zhao
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Tiansong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
145
|
Intestine-specific removal of DAF-2 nearly doubles lifespan in Caenorhabditis elegans with little fitness cost. Nat Commun 2022; 13:6339. [PMID: 36284093 PMCID: PMC9596710 DOI: 10.1038/s41467-022-33850-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
Twenty-nine years following the breakthrough discovery that a single-gene mutation of daf-2 doubles Caenorhabditis elegans lifespan, it remains unclear where this insulin/IGF-1 receptor gene is expressed and where it acts to regulate ageing. Using knock-in fluorescent reporters, we determined that daf-2 and its downstream transcription factor daf-16 are expressed ubiquitously. Using tissue-specific targeted protein degradation, we determined that intracellular DAF-2-to-DAF-16 signaling in the intestine plays a major role in lifespan regulation, while that in the hypodermis, neurons, and germline plays a minor role. Notably, intestine-specific loss of DAF-2 activates DAF-16 in and outside the intestine, causes almost no adverse effects on development and reproduction, and extends lifespan by 94% in a way that partly requires non-intestinal DAF-16. Consistent with intestine supplying nutrients to the entire body, evidence from this and other studies suggests that altered metabolism, particularly down-regulation of protein and RNA synthesis, mediates longevity by reduction of insulin/IGF-1 signaling.
Collapse
|
146
|
Son S, Choi MK, Lim DS, Shim J, Lee J. A genetic screen for aldicarb resistance of Caenorhabditiselegans dauer larvae uncovers 2 alleles of dach-1, a cytochrome P450 gene. G3 (BETHESDA, MD.) 2022; 12:6747961. [PMID: 36194018 PMCID: PMC9713407 DOI: 10.1093/g3journal/jkac266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/26/2022] [Indexed: 12/05/2022]
Abstract
Animals exhibit phenotypic plasticity through the interaction of genes with the environment, and little is known about the genetic factors that change synaptic function at different developmental stages. Here, we investigated the genetic determinants of how animal's sensitivity to drugs that alter synaptic activity is regulated at a specific developmental stage using the free-living nematode Caenorhabditis elegans. C. elegans enters the stress-resistant dauer larval stage under harsh conditions. Although dauer is known to have reduced permeability and increased resistance to most known exogenous chemicals, we discovered that dauer is hypersensitive to a cholinesterase inhibitor, aldicarb. To investigate genes regulating dauer-specific acetylcholine transduction, we first screened for aldicarb-resistant mutations in dauer and then performed a secondary screen to rule out aldicarb-resistant mutations that also affect adults. We isolated 2 different mutations of a single gene called cyp-34A4 or dach-1 encoding a cytochrome P450. In the nondauer stages, dach-1 is mainly expressed in the intestine, but its expression is robustly increased in the epidermis of dauers. By tissue-specific rescue experiments, we found that dach-1 modulates aldicarb sensitivity in a cell nonautonomous manner. In addition, dach-1 plays pleiotropic functions in dauers by regulating quiescence and surviving heat shock and hyperosmolar stress. Our study reveals novel functions of the cytochrome P450 in synaptic and physiological changes during the developmental plasticity.
Collapse
Affiliation(s)
| | | | - Daisy S Lim
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Jaegal Shim
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Junho Lee
- Corresponding author: Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
147
|
Shrivastava A, Sandhof CA, Reinle K, Jawed A, Ruger-Herreros C, Schwarz D, Creamer D, Nussbaum-Krammer C, Mogk A, Bukau B. The cytoprotective sequestration activity of small heat shock proteins is evolutionarily conserved. J Cell Biol 2022; 221:213447. [PMID: 36069810 PMCID: PMC9458469 DOI: 10.1083/jcb.202202149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/21/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
The chaperone-mediated sequestration of misfolded proteins into inclusions is a pivotal cellular strategy to maintain proteostasis in Saccharomyces cerevisiae, executed by small heat shock proteins (sHsps) Hsp42 and Btn2. Direct homologs of Hsp42 and Btn2 are absent in other organisms, questioning whether sequestration represents a conserved proteostasis strategy and, if so, which factors are involved. We examined sHsps from Escherchia coli, Caenorhabditis elegans, and humans for their ability to complement the defects of yeast sequestrase mutants. We show that sequestration of misfolded proteins is an original and widespread activity among sHsps executed by specific family members. Sequestrase positive C. elegans' sHsps harbor specific sequence features, including a high content of aromatic and methionine residues in disordered N-terminal extensions. Those sHsps buffer limitations in Hsp70 capacity in C. elegans WT animals and are upregulated in long-lived daf-2 mutants, contributing to lifespan extension. Cellular protection by sequestration of misfolded proteins is, therefore, an evolutionarily conserved activity of the sHsp family.
Collapse
Affiliation(s)
- Aseem Shrivastava
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kevin Reinle
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Areeb Jawed
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carmen Ruger-Herreros
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Schwarz
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Declan Creamer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
148
|
Milholland B, Vijg J. Why Gilgamesh failed: the mechanistic basis of the limits to human lifespan. NATURE AGING 2022; 2:878-884. [PMID: 37118288 PMCID: PMC11922162 DOI: 10.1038/s43587-022-00291-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/05/2022] [Indexed: 04/30/2023]
Abstract
The purpose of this Perspective is to clarify for an interdisciplinary audience the fundamental concepts of human longevity and provide evidence for a limit to human lifespan. This observed limit is placed into a broader framework by showing how it has arisen through the process of evolution and by enumerating the molecular mechanisms that may enforce it. Finally, we look toward potential future developments and the prospects for possibly circumventing the current limit.
Collapse
Affiliation(s)
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
149
|
Brenner C. Sirtuins are Not Conserved Longevity Genes. LIFE METABOLISM 2022; 1:122-133. [PMID: 37035412 PMCID: PMC10081735 DOI: 10.1093/lifemeta/loac025] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/14/2022]
Abstract
It is central to biology that sequence conservation suggests functional conservation. Animal longevity is an emergent property of selected traits that integrates capacities to perform physical and mental functions after reproductive maturity. Though the yeast SIR2 gene was nominated as a longevity gene based on extended replicative longevity of old mother cells, this is not a selected trait: SIR2 is selected against in chronological aging and the direct targets of SIR2 in replicative lifespan are not conserved. Though it would be difficult to imagine how a gene that advantages 1 in 5 million yeast cells could have anticipated causes of aging in animals, overexpression of SIR2 homologs was tested in invertebrates for longevity. Because artifactual positive results were reported years before they were sorted out and because it was not known that SIR2 functions as a pro-aging gene in yeast chronological aging and in flies subject to amino acid deprivation, a global pursuit of longevity phenotypes was driven by a mixture of framing bias, confirmation bias and hype. Review articles that propagate these biases are so rampant that few investigators have considered how weak the case ever was for sirtuins as longevity genes. Acknowledging that a few positive associations between sirtuins and longevity have been identified after thousands of person-years and billions of dollars of effort, we review the data and suggest rejection of the notions that sirtuins 1) have any specific connection to lifespan in animals and 2) are primary mediators of the beneficial effects of NAD repletion.
Collapse
Affiliation(s)
- Charles Brenner
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010USA
| |
Collapse
|
150
|
Lu R, Chen J, Wang F, Wang L, Liu J, Lin Y. Lysosome Inhibition Reduces Basal and Nutrient-Induced Fat Accumulation in Caenorhabditis elegans. Mol Cells 2022; 45:649-659. [PMID: 36058890 PMCID: PMC9448645 DOI: 10.14348/molcells.2022.0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 11/29/2022] Open
Abstract
A long-term energy nutritional imbalance fundamentally causes the development of obesity and associated fat accumulation. Lysosomes, as nutrient-sensing and lipophagy centers, critically control cellular lipid catabolism in response to nutrient deprivation. However, whether lysosome activity is directly involved in nutrient-induced fat accumulation remains unclear. In this study, worm fat accumulation was induced by 1 mM glucose or 0.02 mM palmitic acid supplementation. Along with the elevation of fat accumulation, lysosomal number and acidification were also increased, suggesting that lysosome activity might be correlated with nutrient-induced fat deposition in Caenorhabditis elegans. Furthermore, treatments with the lysosomal inhibitors chloroquine and leupeptin significantly reduced basal and nutrient-induced fat accumulation in C. elegans. The knockdown of hlh-30, which is a critical gene in lysosomal biogenesis, also resulted in worm fat loss. Finally, the mutation of aak-2, daf-15, and rsks-1 showed that mTORC1 (mechanistic target of rapamycin complex-1) signaling mediated the effects of lysosomes on basal and nutrient-induced fat accumulation in C. elegans. Overall, this study reveals the previously undescribed role of lysosomes in overnutrition sensing, suggesting a new strategy for controlling body fat accumulation.
Collapse
Affiliation(s)
- Rui Lu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|