1
|
Yan J, Bhanshali F, Shuzenji C, Mendenhall TT, Taylor SKB, Ermakova G, Cheng X, Bai P, Diwan G, Seraj D, Meyer JN, Sorensen PH, Hartman JH, Taubert S. Eukaryotic Elongation Factor 2 Kinase EFK-1/eEF2K promotes starvation resistance by preventing oxidative damage in C. elegans. Nat Commun 2025; 16:1752. [PMID: 39966347 PMCID: PMC11836464 DOI: 10.1038/s41467-025-56766-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
Cells and organisms frequently experience starvation. To survive, they mount an evolutionarily conserved stress response. A vital component in the mammalian starvation response is eukaryotic elongation factor 2 (eEF2) kinase (eEF2K), which suppresses translation in starvation by phosphorylating and inactivating the translation elongation driver eEF2. C. elegans EFK-1/eEF2K phosphorylates EEF-2/eEF2 on a conserved residue and is required for starvation survival, but how it promotes survival remains unclear. Surprisingly, we found that eEF2 phosphorylation is unchanged in starved C. elegans and EFK-1's kinase activity is dispensable for starvation survival, suggesting that efk-1 promotes survival via a noncanonical pathway. We show that efk-1 upregulates transcription of DNA repair pathways, nucleotide excision repair (NER) and base excision repair (BER), to promote starvation survival. Furthermore, efk-1 suppresses oxygen consumption and ROS production in starvation to prevent oxidative stress. Thus, efk-1 enables starvation survival by protecting animals from starvation-induced oxidative damage through an EEF-2-independent pathway.
Collapse
Affiliation(s)
- Junran Yan
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Graduate Program in Cell & Developmental Biology, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Forum Bhanshali
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Catalera BioSolutions, 199 W 6th Ave, Vancouver, BC, V5Y 1K3, Canada
| | - Chiaki Shuzenji
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Tsultrim T Mendenhall
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, 29425, USA
| | - Shane K B Taylor
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Glafira Ermakova
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Xuanjin Cheng
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Canada's Michael Smith Genome Sciences Centre, 570 W 7th Ave, Vancouver, BC, V5Z 4S6, Canada
| | - Pamela Bai
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Gahan Diwan
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Biology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Donna Seraj
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, 27708-0328, USA
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, 675 W 10th Ave, Vancouver, BC, V6T 1Z4, Canada
- Department of Molecular Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, 29425, USA
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, 950 W 28 th Ave, Vancouver, BC, V5Z 4H4, Canada.
- Edwin S.H. Leong Centre for Healthy Aging, The University of British Columbia, 117-2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- British Columbia Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada.
- Graduate Program in Cell & Developmental Biology, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada.
- Department of Medical Genetics, The University of British Columbia, 950 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada.
| |
Collapse
|
2
|
Shaulson ED, Cohen AA, Picard M. The brain-body energy conservation model of aging. NATURE AGING 2024; 4:1354-1371. [PMID: 39379694 DOI: 10.1038/s43587-024-00716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
Aging involves seemingly paradoxical changes in energy metabolism. Molecular damage accumulation increases cellular energy expenditure, yet whole-body energy expenditure remains stable or decreases with age. We resolve this apparent contradiction by positioning the brain as the mediator and broker in the organismal energy economy. As somatic tissues accumulate damage over time, costly intracellular stress responses are activated, causing aging or senescent cells to secrete cytokines that convey increased cellular energy demand (hypermetabolism) to the brain. To conserve energy in the face of a shrinking energy budget, the brain deploys energy conservation responses, which suppress low-priority processes, producing fatigue, physical inactivity, blunted sensory capacities, immune alterations and endocrine 'deficits'. We term this cascade the brain-body energy conservation (BEC) model of aging. The BEC outlines (1) the energetic cost of cellular aging, (2) how brain perception of senescence-associated hypermetabolism may drive the phenotypic manifestations of aging and (3) energetic principles underlying the modifiability of aging trajectories by stressors and geroscience interventions.
Collapse
Affiliation(s)
- Evan D Shaulson
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alan A Cohen
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
3
|
Slade L, Etheridge T, Szewczyk NJ. Consolidating multiple evolutionary theories of ageing suggests a need for new approaches to study genetic contributions to ageing decline. Ageing Res Rev 2024; 100:102456. [PMID: 39153601 DOI: 10.1016/j.arr.2024.102456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Understanding mechanisms of ageing remains a complex challenge for biogerontologists, but recent adaptations of evolutionary ageing theories offer a compelling lens in which to view both age-related molecular and physiological deterioration. Ageing is commonly associated with progressive declines in biochemical and molecular processes resulting from damage accumulation, yet the role of continued developmental gene activation is less appreciated. Natural selection pressures are at their highest in youthful periods to modify gene expression towards maximising reproductive capacity. After sexual maturation, selective pressure diminishes, subjecting individuals to maladaptive pleiotropic gene functions that were once beneficial for developmental growth but become pathogenic later in life. Due to this selective 'shadowing' in ageing, mechanisms to counter such hyper/hypofunctional genes are unlikely to evolve. Interventions aimed at targeting gene hyper/hypofunction during ageing might, therefore, represent an attractive therapeutic strategy. The nematode Caenorhabditis elegans offers a strong model for post-reproductive mechanistic and therapeutic investigations, yet studies examining the mechanisms of, and countermeasures against, ageing decline largely intervene from larval stages onwards. Importantly, however, lifespan extending conditions frequently impair early-life fitness and fail to correspondingly increase healthspan. Here, we consolidate multiple evolutionary theories of ageing and discuss data supporting hyper/hypofunctional changes at a global molecular and functional level in C. elegans, and how classical lifespan-extension mutations alter these dynamics. The relevance of such mutant models for exploring mechanisms of ageing are discussed, highlighting that post-reproductive gene optimisation represents a more translatable approach for C. elegans research that is not constrained by evolutionary trade-offs. Where some genetic mutations in C. elegans that promote late-life health map accordingly with healthy ageing in humans, other widely used genetic mutations that extend worm lifespan are associated with life-limiting pathologies in people. Lifespan has also become the gold standard for quantifying 'ageing', but we argue that gerospan compression (i.e., 'healthier' ageing) is an appropriate goal for anti-ageing research, the mechanisms of which appear distinct from those regulating lifespan alone. There is, therefore, an evident need to re-evaluate experimental approaches to study the role of hyper/hypofunctional genes in ageing in C. elegans.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, Exeter, UK.
| | - Timothy Etheridge
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Nathaniel J Szewczyk
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Athens, OH 45701, United States.
| |
Collapse
|
4
|
Mir DA, Ma Z, Horrocks J, Rogers A. Stress-Induced Eukaryotic Translational Regulatory Mechanisms. JOURNAL OF CLINICAL AND MEDICAL SCIENCES 2024; 8:1000277. [PMID: 39364184 PMCID: PMC11448810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The eukaryotic protein synthesis process entails intricate stages governed by diverse mechanisms to tightly regulate translation. Translational regulation during stress is pivotal for maintaining cellular homeostasis, ensuring the accurate expression of essential proteins is important for survival. This selective translational control mechanism is integral to cellular adaptation and resilience under adverse conditions. This review manuscript explores various mechanisms involved in selective translational regulation, focusing on mRNA-specific and global regulatory processes. Key aspects of translational control include translation initiation, which is often a rate-limiting step, and involves the formation of the eIF4F complex and recruitment of mRNA to ribosomes. Regulation of translation initiation factors, such as eIF4E, eIF4E2, and eIF2, through phosphorylation and interactions with binding proteins, modulates translation efficiency under stress conditions. This review also highlights the control of translation initiation through factors like the eIF4F complex and the ternary complex and also underscores the importance of eIF2α phosphorylation in stress granule formation and cellular stress responses. Additionally, the impact of amino acid deprivation, mTOR signaling, and ribosome biogenesis on translation regulation and cellular adaptation to stress is also discussed. Understanding the intricate mechanisms of translational regulation during stress provides insights into cellular adaptation mechanisms and potential therapeutic targets for various diseases, offering valuable avenues for addressing conditions associated with dysregulated protein synthesis.
Collapse
Affiliation(s)
- Dilawar Ahmad Mir
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Maine, United States of America
| | - Zhengxin Ma
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Maine, United States of America
| | - Jordan Horrocks
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Maine, United States of America
| | - Aric Rogers
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Maine, United States of America
| |
Collapse
|
5
|
Mir DA, Ma Z, Horrocks J, Rogers AN. Stress-induced Eukaryotic Translational Regulatory Mechanisms. ARXIV 2024:arXiv:2405.01664v1. [PMID: 38745702 PMCID: PMC11092689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The eukaryotic protein synthesis process entails intricate stages governed by diverse mechanisms to tightly regulate translation. Translational regulation during stress is pivotal for maintaining cellular homeostasis, ensuring the accurate expression of essential proteins crucial for survival. This selective translational control mechanism is integral to cellular adaptation and resilience under adverse conditions. This review manuscript explores various mechanisms involved in selective translational regulation, focusing on mRNA-specific and global regulatory processes. Key aspects of translational control include translation initiation, which is often a rate-limiting step, and involves the formation of the eIF4F complex and recruitment of mRNA to ribosomes. Regulation of translation initiation factors, such as eIF4E, eIF4E2, and eIF2, through phosphorylation and interactions with binding proteins, modulates translation efficiency under stress conditions. This review also highlights the control of translation initiation through factors like the eIF4F complex and the ternary complex and also underscores the importance of eIF2α phosphorylation in stress granule formation and cellular stress responses. Additionally, the impact of amino acid deprivation, mTOR signaling, and ribosome biogenesis on translation regulation and cellular adaptation to stress is also discussed. Understanding the intricate mechanisms of translational regulation during stress provides insights into cellular adaptation mechanisms and potential therapeutic targets for various diseases, offering valuable avenues for addressing conditions associated with dysregulated protein synthesis.
Collapse
Affiliation(s)
- Dilawar Ahmad Mir
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, ME
| | - Zhengxin Ma
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, ME
| | - Jordan Horrocks
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, ME
| | - Aric N Rogers
- Kathryn W. Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, ME
| |
Collapse
|
6
|
En A, Takemoto K, Yamakami Y, Nakabayashi K, Fujii M. Upregulated expression of lamin B receptor increases cell proliferation and suppresses genomic instability: implications for cellular immortalization. FEBS J 2024; 291:2155-2171. [PMID: 38462947 DOI: 10.1111/febs.17113] [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: 06/05/2023] [Revised: 01/04/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
Mammalian somatic cells undergo terminal proliferation arrest after a limited number of cell divisions, a phenomenon termed cellular senescence. However, cells acquire the ability to proliferate infinitely (cellular immortalization) through multiple genetic alterations. Inactivation of tumor suppressor genes such as p53, RB and p16 is important for cellular immortalization, although additional molecular alterations are required for cellular immortalization to occur. Here, we aimed to gain insights into these molecular alterations. Given that cellular immortalization is the escape of cells from cellular senescence, genes that regulate cellular senescence are likely to be involved in cellular immortalization. Because senescent cells show altered heterochromatin organization, we investigated the implications of lamin A/C, lamin B1 and lamin B receptor (LBR), which regulate heterochromatin organization, in cellular immortalization. We employed human immortalized cell lines, KMST-6 and SUSM-1, and found that expression of LBR was upregulated upon cellular immortalization and downregulated upon cellular senescence. In addition, knockdown of LBR induced cellular senescence with altered chromatin configuration. Additionally, enforced expression of LBR increased cell proliferation likely through suppression of genome instability in human primary fibroblasts that expressed the simian virus 40 large T antigen (TAg), which inactivates p53 and RB. Furthermore, expression of TAg or knockdown of p53 led to upregulated LBR expression. These observations suggested that expression of LBR might be upregulated to suppress genome instability in TAg-expressing cells, and, consequently, its upregulated expression assisted the proliferation of TAg-expressing cells (i.e. p53/RB-defective cells). Our findings suggest a crucial role for LBR in the process of cellular immortalization.
Collapse
Affiliation(s)
- Atsuki En
- Graduate School of Nanobioscience, Yokohama City University, Japan
| | - Kentaro Takemoto
- Graduate School of Nanobioscience, Yokohama City University, Japan
| | - Yoshimi Yamakami
- Graduate School of Nanobioscience, Yokohama City University, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo, Japan
| | - Michihiko Fujii
- Graduate School of Nanobioscience, Yokohama City University, Japan
| |
Collapse
|
7
|
Venz R, Goyala A, Soto-Gamez A, Yenice T, Demaria M, Ewald CY. In-vivo screening implicates endoribonuclease Regnase-1 in modulating senescence-associated lysosomal changes. GeroScience 2024; 46:1499-1514. [PMID: 37644339 PMCID: PMC10828269 DOI: 10.1007/s11357-023-00909-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
Accumulation of senescent cells accelerates aging and age-related diseases, whereas preventing this accumulation extends the lifespan in mice. A characteristic of senescent cells is increased staining with β-galactosidase (β-gal) ex vivo. Here, we describe a progressive accumulation of β-gal staining in the model organism C. elegans during aging. We show that distinct pharmacological and genetic interventions targeting the mitochondria and the mTORC1 to the nuclear core complex axis, the non-canonical apoptotic, and lysosomal-autophagy pathways slow the age-dependent accumulation of β-gal. We identify a novel gene, rege-1/Regnase-1/ZC3H12A/MCPIP1, modulating β-gal staining via the transcription factor ets-4/SPDEF. We demonstrate that knocking down Regnase-1 in human cell culture prevents senescence-associated β-gal accumulation. Our data provide a screening pipeline to identify genes and drugs modulating senescence-associated lysosomal phenotypes.
Collapse
Affiliation(s)
- Richard Venz
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Abel Soto-Gamez
- European Institute for the Biology of Aging (ERIBA)/University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Tugce Yenice
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Marco Demaria
- European Institute for the Biology of Aging (ERIBA)/University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland.
| |
Collapse
|
8
|
Sharifi S, Chaudhari P, Martirosyan A, Eberhardt AO, Witt F, Gollowitzer A, Lange L, Woitzat Y, Okoli EM, Li H, Rahnis N, Kirkpatrick J, Werz O, Ori A, Koeberle A, Bierhoff H, Ermolaeva M. Reducing the metabolic burden of rRNA synthesis promotes healthy longevity in Caenorhabditis elegans. Nat Commun 2024; 15:1702. [PMID: 38402241 PMCID: PMC10894287 DOI: 10.1038/s41467-024-46037-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Ribosome biogenesis is initiated by RNA polymerase I (Pol I)-mediated synthesis of pre-ribosomal RNA (pre-rRNA). Pol I activity was previously linked to longevity, but the underlying mechanisms were not studied beyond effects on nucleolar structure and protein translation. Here we use multi-omics and functional tests to show that curtailment of Pol I activity remodels the lipidome and preserves mitochondrial function to promote longevity in Caenorhabditis elegans. Reduced pre-rRNA synthesis improves energy homeostasis and metabolic plasticity also in human primary cells. Conversely, the enhancement of pre-rRNA synthesis boosts growth and neuromuscular performance of young nematodes at the cost of accelerated metabolic decline, mitochondrial stress and premature aging. Moreover, restriction of Pol I activity extends lifespan more potently than direct repression of protein synthesis, and confers geroprotection even when initiated late in life, showcasing this intervention as an effective longevity and metabolic health treatment not limited by aging.
Collapse
Affiliation(s)
- Samim Sharifi
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Matter Bio, Inc., Brooklyn, NY, 11237, USA
| | - Prerana Chaudhari
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Asya Martirosyan
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Joseph-Stelzmann-Straße 26, 50931, Cologne, Germany
| | - Alexander Otto Eberhardt
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lisa Lange
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Yvonne Woitzat
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | | | - Huahui Li
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, PR China
| | - Norman Rahnis
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Joanna Kirkpatrick
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
- Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Holger Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, Jena, 07745, Germany.
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
9
|
Clay KJ, Yang Y, Clark C, Petrascheck M. Proteostasis is differentially modulated by inhibition of translation initiation or elongation. eLife 2023; 12:e76465. [PMID: 37795690 PMCID: PMC10581687 DOI: 10.7554/elife.76465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
Recent work has revealed an increasingly important role for mRNA translation in maintaining proteostasis. Here, we use chemical inhibitors targeting discrete steps of translation to compare how lowering the concentration of all or only translation initiation-dependent proteins rescues Caenorhabditis elegans from proteotoxic stress. We systematically challenge proteostasis and show that pharmacologically inhibiting translation initiation or elongation elicits a distinct protective profile. Inhibiting elongation protects from heat and proteasome dysfunction independently from HSF-1 but does not protect from age-associated protein aggregation. Conversely, inhibition of initiation protects from heat and age-associated protein aggregation and increases lifespan, dependent on hsf-1, but does not protect from proteotoxicity caused by proteasome dysfunction. Surprisingly, we find that the ability of the translation initiation machinery to control the concentration of newly synthesized proteins depends on HSF-1. Inhibition of translation initiation in wild-type animals reduces the concentration of newly synthesized proteins but increases it in hsf-1 mutants. Our findings suggest that the HSF-1 pathway is not only a downstream target of translation but also directly cooperates with the translation initiation machinery to control the concentration of newly synthesized proteins to restore proteostasis.
Collapse
Affiliation(s)
- Khalyd J Clay
- Department of Molecular Medicine, Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Yongzhi Yang
- Department of Molecular Medicine, Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Christina Clark
- Department of Molecular Medicine, Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Michael Petrascheck
- Department of Molecular Medicine, Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| |
Collapse
|
10
|
Kim HS, Parker DJ, Hardiman MM, Munkácsy E, Jiang N, Rogers AN, Bai Y, Brent C, Mobley JA, Austad SN, Pickering AM. Early-adulthood spike in protein translation drives aging via juvenile hormone/germline signaling. Nat Commun 2023; 14:5021. [PMID: 37596266 PMCID: PMC10439225 DOI: 10.1038/s41467-023-40618-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/01/2023] [Indexed: 08/20/2023] Open
Abstract
Protein translation (PT) declines with age in invertebrates, rodents, and humans. It has been assumed that elevated PT at young ages is beneficial to health and PT ends up dropping as a passive byproduct of aging. In Drosophila, we show that a transient elevation in PT during early-adulthood exerts long-lasting negative impacts on aging trajectories and proteostasis in later-life. Blocking the early-life PT elevation robustly improves life-/health-span and prevents age-related protein aggregation, whereas transiently inducing an early-life PT surge in long-lived fly strains abolishes their longevity/proteostasis benefits. The early-life PT elevation triggers proteostatic dysfunction, silences stress responses, and drives age-related functional decline via juvenile hormone-lipid transfer protein axis and germline signaling. Our findings suggest that PT is adaptively suppressed after early-adulthood, alleviating later-life proteostatic burden, slowing down age-related functional decline, and improving lifespan. Our work provides a theoretical framework for understanding how lifetime PT dynamics shape future aging trajectories.
Collapse
Affiliation(s)
- Harper S Kim
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Medical Scientist Training Program, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Danitra J Parker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, 77030, USA
| | - Madison M Hardiman
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erin Munkácsy
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Nisi Jiang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Aric N Rogers
- MDI Biological Laboratory, Bar Harbor, ME, 04672, USA
| | - Yidong Bai
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Colin Brent
- USDA-ARS Arid Land Agricultural Research Center, Maricopa, AZ, 85138, USA
| | - James A Mobley
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, 35249, USA
| | - Steven N Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Nathan Shock Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Andrew M Pickering
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, 77030, USA.
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
11
|
Vijay Kumar MJ, Morales R, Tsvetkov AS. G-quadruplexes and associated proteins in aging and Alzheimer's disease. FRONTIERS IN AGING 2023; 4:1164057. [PMID: 37323535 PMCID: PMC10267416 DOI: 10.3389/fragi.2023.1164057] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Aging is a prominent risk factor for many neurodegenerative disorders, such as Alzheimer's disease (AD). Alzheimer's disease is characterized by progressive cognitive decline, memory loss, and neuropsychiatric and behavioral symptoms, accounting for most of the reported dementia cases. This disease is now becoming a major challenge and burden on modern society, especially with the aging population. Over the last few decades, a significant understanding of the pathophysiology of AD has been gained by studying amyloid deposition, hyperphosphorylated tau, synaptic dysfunction, oxidative stress, calcium dysregulation, and neuroinflammation. This review focuses on the role of non-canonical secondary structures of DNA/RNA G-quadruplexes (G4s, G4-DNA, and G4-RNA), G4-binding proteins (G4BPs), and helicases, and their roles in aging and AD. Being critically important for cellular function, G4s are involved in the regulation of DNA and RNA processes, such as replication, transcription, translation, RNA localization, and degradation. Recent studies have also highlighted G4-DNA's roles in inducing DNA double-strand breaks that cause genomic instability and G4-RNA's participation in regulating stress granule formation. This review emphasizes the significance of G4s in aging processes and how their homeostatic imbalance may contribute to the pathophysiology of AD.
Collapse
Affiliation(s)
- M. J. Vijay Kumar
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| | - Rodrigo Morales
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Andrey S. Tsvetkov
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
12
|
Kim HS, Pickering AM. Protein translation paradox: Implications in translational regulation of aging. Front Cell Dev Biol 2023; 11:1129281. [PMID: 36711035 PMCID: PMC9880214 DOI: 10.3389/fcell.2023.1129281] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Protein translation is an essential cellular process playing key roles in growth and development. Protein translation declines over the course of age in multiple animal species, including nematodes, fruit flies, mice, rats, and even humans. In all these species, protein translation transiently peaks in early adulthood with a subsequent drop over the course of age. Conversely, lifelong reductions in protein translation have been found to extend lifespan and healthspan in multiple animal models. These findings raise the protein synthesis paradox: age-related declines in protein synthesis should be detrimental, but life-long reductions in protein translation paradoxically slow down aging and prolong lifespan. This article discusses the nature of this paradox and complies an extensive body of work demonstrating protein translation as a modulator of lifespan and healthspan.
Collapse
Affiliation(s)
- Harper S. Kim
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Medical Scientist Training Program, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew M. Pickering
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
13
|
Sturm G, Karan KR, Monzel AS, Santhanam B, Taivassalo T, Bris C, Ware SA, Cross M, Towheed A, Higgins-Chen A, McManus MJ, Cardenas A, Lin J, Epel ES, Rahman S, Vissing J, Grassi B, Levine M, Horvath S, Haller RG, Lenaers G, Wallace DC, St-Onge MP, Tavazoie S, Procaccio V, Kaufman BA, Seifert EL, Hirano M, Picard M. OxPhos defects cause hypermetabolism and reduce lifespan in cells and in patients with mitochondrial diseases. Commun Biol 2023; 6:22. [PMID: 36635485 PMCID: PMC9837150 DOI: 10.1038/s42003-022-04303-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/26/2022] [Indexed: 01/13/2023] Open
Abstract
Patients with primary mitochondrial oxidative phosphorylation (OxPhos) defects present with fatigue and multi-system disorders, are often lean, and die prematurely, but the mechanistic basis for this clinical picture remains unclear. By integrating data from 17 cohorts of patients with mitochondrial diseases (n = 690) we find evidence that these disorders increase resting energy expenditure, a state termed hypermetabolism. We examine this phenomenon longitudinally in patient-derived fibroblasts from multiple donors. Genetically or pharmacologically disrupting OxPhos approximately doubles cellular energy expenditure. This cell-autonomous state of hypermetabolism occurs despite near-normal OxPhos coupling efficiency, excluding uncoupling as a general mechanism. Instead, hypermetabolism is associated with mitochondrial DNA instability, activation of the integrated stress response (ISR), and increased extracellular secretion of age-related cytokines and metabokines including GDF15. In parallel, OxPhos defects accelerate telomere erosion and epigenetic aging per cell division, consistent with evidence that excess energy expenditure accelerates biological aging. To explore potential mechanisms for these effects, we generate a longitudinal RNASeq and DNA methylation resource dataset, which reveals conserved, energetically demanding, genome-wide recalibrations. Taken together, these findings highlight the need to understand how OxPhos defects influence the energetic cost of living, and the link between hypermetabolism and aging in cells and patients with mitochondrial diseases.
Collapse
Affiliation(s)
- Gabriel Sturm
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Kalpita R Karan
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Balaji Santhanam
- Departments of Biological Sciences, Systems Biology, and Biochemistry and Molecular Biophysics, Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Tanja Taivassalo
- Department of Physiology and Functional Genomics, Clinical and Translational Research Building, University of Florida, Gainesville, FL, USA
| | - Céline Bris
- Department of Genetics and Neurology, Angers Hospital, Angers, France
- UMR CNRS 6015, INSERM U1083, MITOVASC, SFR ICAT, Université d'Angers, Angers, France
| | - Sarah A Ware
- Department of Medicine, Vascular Medicine Institute and Center for Metabolic and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marissa Cross
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Atif Towheed
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Internal Medicine-Pediatrics Residency Program, University of Pittsburgh Medical Centre, Pittsburgh, PA, USA
| | - Albert Higgins-Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Meagan J McManus
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Elissa S Epel
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Shamima Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, and Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bruno Grassi
- Department of Medicine, University of Udine, Udine, Italy
| | | | | | - Ronald G Haller
- Neuromuscular Center, Institute for Exercise and Environmental Medicine of Texas Health Resources and Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guy Lenaers
- Department of Genetics and Neurology, Angers Hospital, Angers, France
- UMR CNRS 6015, INSERM U1083, MITOVASC, SFR ICAT, Université d'Angers, Angers, France
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marie-Pierre St-Onge
- Center of Excellence for Sleep & Circadian Research and Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Saeed Tavazoie
- Departments of Biological Sciences, Systems Biology, and Biochemistry and Molecular Biophysics, Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Vincent Procaccio
- Department of Genetics and Neurology, Angers Hospital, Angers, France
- UMR CNRS 6015, INSERM U1083, MITOVASC, SFR ICAT, Université d'Angers, Angers, France
| | - Brett A Kaufman
- Department of Medicine, Vascular Medicine Institute and Center for Metabolic and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erin L Seifert
- Department of Pathology and Genomic Medicine, and MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
14
|
Wu G, Xu J, Wang Q, Fang Z, Fang Y, Jiang Y, Zhang X, Cheng X, Sun J, Le G. Methionine-Restricted Diet: A Feasible Strategy Against Chronic or Aging-Related Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5-19. [PMID: 36571820 DOI: 10.1021/acs.jafc.2c05829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Dietary methionine restriction (MR) has been associated with multifaceted health-promoting effects. MR is conducive to prevention of several chronic diseases and cancer, and extension of lifespan. A growing number of studies on new phenotypes and mechanisms of MR have become available in the past five years, especially in angiogenesis, neurodegenerative diseases, intestinal microbiota, and intestinal barrier function. In this review, we summarize the characteristics and advantages of MR, and current knowledge on the physiological responses and effects of MR on chronic diseases and aging-associated pathologies. Potential mechanisms, in which hydrogen sulfide, fibroblast growth factor 21, gut microbiota, short-chain fatty acids, and so on are involved, are discussed. Moreover, directions for epigenetics and gut microbiota in an MR diet are presented in future perspectives. This review comprehensively summarizes the novel roles and interpretations of the mechanisms underlying MR in the prevention of chronic diseases and aging.
Collapse
Affiliation(s)
- Guoqing Wu
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jingxuan Xu
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Qiyao Wang
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Ziyang Fang
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yucheng Fang
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yujie Jiang
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiaohong Zhang
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiangrong Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266021, China
| | - Guowei Le
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
15
|
Diwan B, Sharma R. Nutritional components as mitigators of cellular senescence in organismal aging: a comprehensive review. Food Sci Biotechnol 2022; 31:1089-1109. [PMID: 35756719 PMCID: PMC9206104 DOI: 10.1007/s10068-022-01114-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 11/06/2022] Open
Abstract
The process of cellular senescence is rapidly emerging as a modulator of organismal aging and disease. Targeting the development and removal of senescent cells is considered a viable approach to achieving improved organismal healthspan and lifespan. Nutrition and health are intimately linked and an appropriate dietary regimen can greatly impact organismal response to stress and diseases including during aging. With a renewed focus on cellular senescence, emerging studies demonstrate that both primary and secondary nutritional elements such as carbohydrates, proteins, fatty acids, vitamins, minerals, polyphenols, and probiotics can influence multiple aspects of cellular senescence. The present review describes the recent molecular aspects of cellular senescence-mediated understanding of aging and then studies available evidence of the cellular senescence modulatory attributes of major and minor dietary elements. Underlying pathways and future research directions are deliberated to promote a nutrition-centric approach for targeting cellular senescence and thus improving human health and longevity.
Collapse
Affiliation(s)
- Bhawna Diwan
- Faculty of Applied Sciences & Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, 173229 India
| | - Rohit Sharma
- Faculty of Applied Sciences & Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, 173229 India
| |
Collapse
|
16
|
Targeting tumor cell senescence and polyploidy as potential therapeutic strategies. Semin Cancer Biol 2022; 81:37-47. [PMID: 33358748 PMCID: PMC8214633 DOI: 10.1016/j.semcancer.2020.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 01/14/2023]
Abstract
Senescence is a unique state of growth arrest that develops in response to a plethora of cellular stresses, including replicative exhaustion, oxidative injury, and genotoxic insults. Senescence has been implicated in the pathogenesis of multiple aging-related pathologies, including cancer. In cancer, senescence plays a dual role, initially acting as a barrier against tumor progression by enforcing a durable growth arrest in premalignant cells, but potentially promoting malignant transformation in neighboring cells through the secretion of pro-tumorigenic drivers. Moreover, senescence is induced in tumor cells upon exposure to a wide variety of conventional and targeted anticancer drugs (termed Therapy-Induced Senescence-TIS), representing a critical contributing factor to therapeutic outcomes. As with replicative or oxidative senescence, TIS manifests as a complex phenotype of macromolecular damage, energetic dysregulation, and altered gene expression. Senescent cells are also frequently polyploid. In vitro studies have suggested that polyploidy may confer upon senescent tumor cells the ability to escape from growth arrest, thereby providing an additional avenue whereby tumor cells escape the lethality of anticancer treatment. Polyploidy in tumor cells is also associated with persistent energy production, chromatin remodeling, self-renewal, stemness and drug resistance - features that are also associated with escape from senescence and conversion to a more malignant phenotype. However, senescent cells are highly heterogenous and can present with variable phenotypes, where polyploidy is one component of a complex reversion process. Lastly, emerging efforts to pharmacologically target polyploid tumor cells might pave the way towards the identification of novel targets for the elimination of senescent tumor cells by the incorporation of senolytic agents into cancer therapeutic strategies.
Collapse
|
17
|
Sahabi S, Jafari-Gharabaghlou D, Zarghami N. A new insight into cell biological and biochemical changes through aging. Acta Histochem 2022; 124:151841. [PMID: 34995929 DOI: 10.1016/j.acthis.2021.151841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022]
Abstract
After several years of extensive research, the main cause of aging is yet elusive. There are some theories about aging, such as stem cell aging, senescent cells accumulation, and neuro-endocrine theories. None of them is able to explain all changes that happen in cells and body through aging. By finding out the main cause of aging, it will be much easier to control, prevent and even reverse the aging process. Our cells, regardless of their replicative capacity, get old through aging and they have almost the same epigenetic age. Different cell signaling pathways contribute to aging. The most important one is mTORC1 that becomes hyperactive in cells that undergo aging. Other significant changes with age are lysosome accumulation, impaired autophagy, and mitophagy. Immune system undergoes gradual changes through aging including a shift from lymphoid to myeloid lineage production as well as increased IL-6 and TNF-α which lead to age-related weight loss and meta-inflammation. Additionally, our endocrine system also experiences some changes that should be taken into consideration when looking for the main cause of aging in the human body. In this review, we planned to summarize some of the changes that happen in cells and the body through aging.
Collapse
|
18
|
Chen L, Liao F, Wu J, Wang Z, Jiang Z, Zhang C, Luo P, Ma L, Gong Q, Wang Y, Wang Q, Luo M, Yang Z, Han S, Shi C. Acceleration of ageing via disturbing mTOR-regulated proteostasis by a new ageing-associated gene PC4. Aging Cell 2021; 20:e13370. [PMID: 33957702 PMCID: PMC8208792 DOI: 10.1111/acel.13370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/21/2021] [Accepted: 03/31/2021] [Indexed: 01/08/2023] Open
Abstract
Research on ageing‐associated genes is important for investigating ageing and anti‐ageing strategies. Here, we firstly reported that the human positive cofactor 4 (PC4), a multifunctional and highly conserved nucleoprotein, is accumulated and activated during ageing and causes global accelerated ageing process by disrupting proteostasis. Mechanistically, PC4 interacts with Sin3‐HDAC complex and inhibits its deacetylated activity, leads to hyper‐acetylation of the histones at the promoters of mTOR‐related genes and causes mTOR signalling activation. Accordingly, mTOR activation causes excessive protein synthesis, resulting in impaired proteostasis and accelerated senescence. These results reveal a new biological function of PC4 in vivo, recognizes PC4 as a new ageing‐associated gene and provides a genetically engineered mouse model to simulate natural ageing. More importantly, our findings also indicate that PC4 is involved in histone acetylation and serves as a potential target to improve proteostasis and delay ageing.
Collapse
Affiliation(s)
- Long Chen
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Fengying Liao
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Jie Wu
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Ziwen Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
- Department of Cardiology Geriatric Cardiovascular Disease Research and Treatment Center 252 Hospital of PLA (82nd Group Army Hospital of PLA) Baoding China
| | - Zhongyong Jiang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Chi Zhang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Peng Luo
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Le Ma
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Qiang Gong
- Department of Hematology Southwest Hospital Third Military Medical University Chongqing China
| | - Yang Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Qing Wang
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Min Luo
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| | - Zeyu Yang
- Breast and Thyroid Surgical Department Chongqing General Hospital University of Chinese Academy of Sciences Chongqing China
| | - Shiqian Han
- Institute of Tropical Medicine Third Military Medical University Chongqing China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University Chongqing China
| |
Collapse
|
19
|
Alessio N, Aprile D, Cappabianca S, Peluso G, Di Bernardo G, Galderisi U. Different Stages of Quiescence, Senescence, and Cell Stress Identified by Molecular Algorithm Based on the Expression of Ki67, RPS6, and Beta-Galactosidase Activity. Int J Mol Sci 2021; 22:3102. [PMID: 33803589 PMCID: PMC8002939 DOI: 10.3390/ijms22063102] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
During their life span, cells have two possible states: a non-cycling, quiescent state (G0) and a cycling, activated state. Cells may enter a reversible G0 state of quiescence or, alternatively, they may undergo an irreversible G0 state. The latter may be a physiological differentiation or, following a stress event, a senescent status. Discrimination among the several G0 states represents a significant investigation, since quiescence, differentiation, and senescence are progressive phenomena with intermediate transitional stages. We used the expression of Ki67, RPS6, and beta-galactosidase to identify healthy cells that progressively enter and leave quiescence through G0-entry, G0 and G0-alert states. We then evaluated how cells may enter senescence following a genotoxic stressful event. We identified an initial stress stage with the expression of beta-galactosidase and Ki67 proliferation marker. Cells may recover from stress events or become senescent passing through early and late senescence states. Discrimination between quiescence and senescence was based on the expression of RPS6, a marker of active protein synthesis that is present in senescent cells but absent in quiescent cells. Even taking into account that fixed G0 states do not exist, our molecular algorithm may represent a method for identifying turning points of G0 transitional states that continuously change.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (N.A.); (D.A.); (G.D.B.)
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (N.A.); (D.A.); (G.D.B.)
| | - Salvatore Cappabianca
- Department of Precision Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy;
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (N.A.); (D.A.); (G.D.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (N.A.); (D.A.); (G.D.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri 38280, Turkey
| |
Collapse
|
20
|
Wang S, Meyer DH, Schumacher B. H3K4me2 regulates the recovery of protein biosynthesis and homeostasis following DNA damage. Nat Struct Mol Biol 2020; 27:1165-1177. [DOI: 10.1038/s41594-020-00513-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 09/02/2020] [Indexed: 01/08/2023]
|
21
|
Bulterijs S, Braeckman BP. Phenotypic Screening in C. elegans as a Tool for the Discovery of New Geroprotective Drugs. Pharmaceuticals (Basel) 2020; 13:E164. [PMID: 32722365 PMCID: PMC7463874 DOI: 10.3390/ph13080164] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023] Open
Abstract
Population aging is one of the largest challenges of the 21st century. As more people live to advanced ages, the prevalence of age-related diseases and disabilities will increase placing an ever larger burden on our healthcare system. A potential solution to this conundrum is to develop treatments that prevent, delay or reduce the severity of age-related diseases by decreasing the rate of the aging process. This ambition has been accomplished in model organisms through dietary, genetic and pharmacological interventions. The pharmacological approaches hold the greatest opportunity for successful translation to the clinic. The discovery of such pharmacological interventions in aging requires high-throughput screening strategies. However, the majority of screens performed for geroprotective drugs in C. elegans so far are rather low throughput. Therefore, the development of high-throughput screening strategies is of utmost importance.
Collapse
Affiliation(s)
- Sven Bulterijs
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, 9000 Ghent, Belgium
| | - Bart P. Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
22
|
Progerin Expression Induces Inflammation, Oxidative Stress and Senescence in Human Coronary Endothelial Cells. Cells 2020; 9:cells9051201. [PMID: 32408587 PMCID: PMC7290406 DOI: 10.3390/cells9051201] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/06/2020] [Accepted: 05/09/2020] [Indexed: 12/16/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare premature aging disorder notably characterized by precocious and deadly atherosclerosis. Almost 90% of HGPS patients carry a LMNA p.G608G splice variant that leads to the expression of a permanently farnesylated abnormal form of prelamin-A, referred to as progerin. Endothelial dysfunction is a key determinant of atherosclerosis, notably during aging. Previous studies have shown that progerin accumulates in HGPS patients’ endothelial cells but also during vascular physiological aging. However, whether progerin expression in human endothelial cells can recapitulate features of endothelial dysfunction is currently unknown. Herein, we evaluated the direct impact of exogenously expressed progerin and wild-type lamin-A on human endothelial cell function and senescence. Our data demonstrate that progerin, but not wild-type lamin-A, overexpression induces endothelial cell dysfunction, characterized by increased inflammation and oxidative stress together with persistent DNA damage, increased cell cycle arrest protein expression and cellular senescence. Inhibition of progerin prenylation using a pravastatin–zoledronate combination partly prevents these defects. Our data suggest a direct proatherogenic role of progerin in human endothelial cells, which could contribute to HGPS-associated early atherosclerosis and also potentially be involved in physiological endothelial aging participating to age-related cardiometabolic diseases.
Collapse
|
23
|
En A, Takauji Y, Ayusawa D, Fujii M. The role of lamin B receptor in the regulation of senescence-associated secretory phenotype (SASP). Exp Cell Res 2020; 390:111927. [PMID: 32126237 DOI: 10.1016/j.yexcr.2020.111927] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Cellular senescence is a phenomenon of irreversible growth arrest of mammalian somatic cells. Senescent cells increase the production of secretory proteins such as inflammatory cytokines, a phenomenon termed senescence-associated secretory phenotype (SASP). SASP is known to have profound effects on organismal health and aging; however, the molecular mechanisms of SASP are not precisely understood. In our previous studies, we have shown that senescent cells show decreased function of lamin B receptor (LBR), a nuclear membrane protein that regulates heterochromatin organization. Here we examined the implication of LBR in the regulation of SASP because senescent cells show altered heterochromatin organization, which would affect gene expression. We found that knock-down of LBR up-regulated the expression of the SASP factors such as IL-6, IL-8, and MMP1 in HeLa cells, even though cellular senescence was not induced by LBR knock-down. Conversely, enforced expression of LBR suppressed their up-regulated expression in senescent cells induced by excess thymidine. Further, our gene expression profile analysis also showed that many secretory proteins were up-regulated by LBR knock-down. We then analyzed the regulatory mechanisms of the expression of SASP factors by LBR, and found that the promoters of these SASP factors associated with LBR in normally growing cells, but dissociated from it in senescent cells. Additionally, we found that enforced expression of LBR decreased the generation of cytoplasmic DNA, which could be involved in SASP, in senescent cells. These findings suggested that LBR would play crucial roles in the regulation of SASP.
Collapse
Affiliation(s)
- Atsuki En
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan
| | - Yuki Takauji
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, 231-0048, Japan
| | - Dai Ayusawa
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, 231-0048, Japan
| | - Michihiko Fujii
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan.
| |
Collapse
|
24
|
En A, Takauji Y, Miki K, Ayusawa D, Fujii M. Lamin B receptor plays a key role in cellular senescence induced by inhibition of the proteasome. FEBS Open Bio 2020; 10:237-250. [PMID: 31825172 PMCID: PMC6996348 DOI: 10.1002/2211-5463.12775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/07/2019] [Accepted: 12/09/2019] [Indexed: 12/04/2022] Open
Abstract
Cellular senescence is a terminal growth arrest phenomenon in mammalian cells. Coordinated regulation of protein synthesis and degradation is required to maintain protein homeostasis in cells; however, senescent cells exhibit decreased activity of the proteasome, a major cellular proteolytic machinery, with an accumulation of proteins. Indeed, we showed that MG132, a proteasome inhibitor, induced cellular senescence through an accumulation of proteins in human cells. We then investigated the mechanisms of cellular senescence induced by protein accumulation by treating cells with MG132. We found that lamin B receptor (LBR), a nuclear membrane protein that regulates heterochromatin organization, was mislocalized and down-regulated in cells on treatment with MG132. Importantly, enforced expression of LBR suppressed cellular senescence induced by MG132. We also showed that LBR was involved in the regulation of chromatin organization in senescent cells, and that endoplasmic reticulum stress and autophagy were likely to be involved in the mislocalization and down-regulation of LBR. These findings indicate that decreased LBR function was responsible for the induction of cellular senescence by MG132, and thus suggest that protein accumulation caused by inhibition of the proteasome induced cellular senescence probably through chromatin dysregulation in human cells.
Collapse
Affiliation(s)
- Atsuki En
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Yuki Takauji
- Graduate School of NanobioscienceYokohama City UniversityJapan
- Ichiban Life CorporationYokohamaJapan
| | - Kensuke Miki
- Graduate School of NanobioscienceYokohama City UniversityJapan
- Ichiban Life CorporationYokohamaJapan
| | - Dai Ayusawa
- Graduate School of NanobioscienceYokohama City UniversityJapan
- Ichiban Life CorporationYokohamaJapan
| | - Michihiko Fujii
- Graduate School of NanobioscienceYokohama City UniversityJapan
| |
Collapse
|
25
|
Ishikawa S, Ishikawa F. Proteostasis failure and cellular senescence in long-term cultured postmitotic rat neurons. Aging Cell 2020; 19:e13071. [PMID: 31762159 PMCID: PMC6974705 DOI: 10.1111/acel.13071] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/05/2019] [Accepted: 10/29/2019] [Indexed: 12/31/2022] Open
Abstract
Cellular senescence, a stress-induced irreversible cell cycle arrest, has been defined for mitotic cells and is implicated in aging of replicative tissues. Age-related functional decline in the brain is often attributed to a failure of protein homeostasis (proteostasis), largely in postmitotic neurons, which accordingly is a process distinct by definition from senescence. It is nevertheless possible that proteostasis failure and cellular senescence have overlapping molecular mechanisms. Here, we identify postmitotic cellular senescence as an adaptive stress response to proteostasis failure. Primary rat hippocampal neurons in long-term cultures show molecular changes indicative of both senescence (senescence-associated β-galactosidase, p16, and loss of lamin B1) and proteostasis failure relevant to Alzheimer's disease. In addition, we demonstrate that the senescent neurons exhibit resistance to stress. Importantly, treatment of the cultures with an mTOR antagonist, protein synthesis inhibitor, or chemical compound that reduces the amount of protein aggregates relieved the proteotoxic stresses as well as the appearance of senescence markers. Our data propose mechanistic insights into the pathophysiological brain aging by establishing senescence as a primary cell-autonomous neuroprotective response.
Collapse
Affiliation(s)
- Shoma Ishikawa
- Department of Gene Mechanisms Graduate School of Biostudies Kyoto University Kyoto Japan
| | - Fuyuki Ishikawa
- Department of Gene Mechanisms Graduate School of Biostudies Kyoto University Kyoto Japan
| |
Collapse
|
26
|
Arai R, En A, Takauji Y, Maki K, Miki K, Fujii M, Ayusawa D. Lamin B receptor (LBR) is involved in the induction of cellular senescence in human cells. Mech Ageing Dev 2019; 178:25-32. [PMID: 30615890 DOI: 10.1016/j.mad.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 11/19/2018] [Accepted: 01/03/2019] [Indexed: 12/13/2022]
Abstract
Cellular senescence is a phenomenon of irreversible growth arrest in mammalian somatic cells in culture. Various stresses induce cellular senescence and indeed, we have found that excess thymidine effectively induces cellular senescence in human cells. Further, many reports indicate the implication of chromatin proteins in cellular senescence. Here we analysed the role of lamin B receptor (LBR), a nuclear envelope protein that regulates heterochromatin organization, in cellular senescence induced by excess thymidine. We then found that the LBR protein was down-regulated and showed aberrant localization in cells upon induction of cellular senescence by excess thymidine. Additionally, we also found that knock-down of LBR facilitated the induction of cellular senescence by excess thymidine in cancerous HeLa cells, and importantly, it induced cellular senescence in normal human diploid fibroblast TIG-7 cells. These results suggested that decreased LBR function is involved in the induction of cellular senescence in human cells.
Collapse
Affiliation(s)
- Rumi Arai
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan
| | - Atsuki En
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan
| | - Yuki Takauji
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, 231-0048, Japan
| | - Keisuke Maki
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan
| | - Kensuke Miki
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, 231-0048, Japan
| | - Michihiko Fujii
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan.
| | - Dai Ayusawa
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, 231-0048, Japan
| |
Collapse
|
27
|
Wang Y, Liu J, Ma X, Cui C, Deenik PR, Henderson PKP, Sigler AL, Cui L. Real-time imaging of senescence in tumors with DNA damage. Sci Rep 2019; 9:2102. [PMID: 30765819 PMCID: PMC6375927 DOI: 10.1038/s41598-019-38511-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/31/2018] [Indexed: 12/22/2022] Open
Abstract
Detection of cellular senescence is important not only in the study of senescence in various biological systems, but also in various practical applications such as image-guided surgical removal of senescent cells, as well as the monitoring of drug-responsiveness during cancer therapies. Due to the lack of suitable imaging probes for senescence detection, particularly in living subjects, we have developed an activatable near-infrared (NIR) molecular probe with far-red excitation, NIR emission, and high "turn-on" ratio upon senescence-associated β-galactosidase (SABG) activation. We present here the first successful demonstration of NIR imaging of DNA damage-induced senescence both in vitro and in human tumor xenograft models.
Collapse
Affiliation(s)
- Ying Wang
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Jun Liu
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Xiaowei Ma
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Chao Cui
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Philip R Deenik
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Paul K P Henderson
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ashton L Sigler
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA
- Departments of Biology and Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Lina Cui
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA.
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87131, USA.
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
28
|
Wu G, Wang Y, Yang Y, Shi Y, Sun J, Xu Y, Luo T, Le G. Dietary Methionine Restriction Upregulates Endogenous H 2 S via miR-328-3p: A Potential Mechanism to Improve Liver Protein Metabolism Efficiency in a Mouse Model of High-fat-diet-induced Obesity. Mol Nutr Food Res 2018; 63:e1800735. [PMID: 30512228 DOI: 10.1002/mnfr.201800735] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/27/2018] [Indexed: 12/21/2022]
Abstract
SCOPE Dietary methionine restriction (MR) promotes multifaceted health benefits. Moreover, lower rate of protein synthesis by dietary MR is associated with life span extension. The goal of this work is to explore how dietary MR would affect protein metabolism in a mouse model of high-fat-diet-induced obesity (DIO). METHODS AND RESULTS DIO mice (male C57BL/6) are subjected to dietary MR for 22 weeks. High-throughput sequencing technology, qRT-PCR analysis, and the dual luciferase reporter assay are performed to verify that MiR-328-3p directly targets cystathionine γ-lyase (CSE) to modulate endogenous H2 S production. Moreover, indicators of endogenous H2 S, fractional synthesis rate (FSR), fractional growth rate (FGR), fractional degradation rate (FDR), and protein retention efficiency (PRE) are analyzed. MR results in an increase in endogenous H2 S to relieve oxidative stress and ER stress to improve protein homeostasis and metabolic efficiency in DIO mice. CONCLUSION Results show that dietary MR increases endogenous H2 S production via miR-328-3p. Furthermore, these results suggest the potential involvement of endogenous H2 S on the efficiency of protein metabolism in dietary MR.
Collapse
Affiliation(s)
- Guoqing Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Yanan Wang
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Yuhui Yang
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Jin Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Yunchong Xu
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Tingyu Luo
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| |
Collapse
|
29
|
Denzel MS, Lapierre LR, Mack HID. Emerging topics in C. elegans aging research: Transcriptional regulation, stress response and epigenetics. Mech Ageing Dev 2018; 177:4-21. [PMID: 30134144 PMCID: PMC6696993 DOI: 10.1016/j.mad.2018.08.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/13/2022]
Abstract
Key discoveries in aging research have been made possible with the use of model organisms. Caenorhabditis elegans is a short-lived nematode that has become a well-established system to study aging. The practicality and powerful genetic manipulations associated with this metazoan have revolutionized our ability to understand how organisms age. 25 years after the publication of the discovery of the daf-2 gene as a genetic modifier of lifespan, C. elegans remains as relevant as ever in the quest to understand the process of aging. Nematode aging research has proven useful in identifying transcriptional regulators, small molecule signals, cellular mechanisms, epigenetic modifications associated with stress resistance and longevity, and lifespan-extending compounds. Here, we review recent discoveries and selected topics that have emerged in aging research using this incredible little worm.
Collapse
Affiliation(s)
- Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| | | |
Collapse
|
30
|
Hu Z, Xia B, Postnikoff SD, Shen ZJ, Tomoiaga AS, Harkness TA, Seol JH, Li W, Chen K, Tyler JK. Ssd1 and Gcn2 suppress global translation efficiency in replicatively aged yeast while their activation extends lifespan. eLife 2018; 7:35551. [PMID: 30117416 PMCID: PMC6097839 DOI: 10.7554/elife.35551] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
Translational efficiency correlates with longevity, yet its role in lifespan determination remains unclear. Using ribosome profiling, translation efficiency is globally reduced during replicative aging in budding yeast by at least two mechanisms: Firstly, Ssd1 is induced during aging, sequestering mRNAs to P-bodies. Furthermore, Ssd1 overexpression in young cells reduced translation and extended lifespan, while loss of Ssd1 reduced the translational deficit of old cells and shortened lifespan. Secondly, phosphorylation of eIF2α, mediated by the stress kinase Gcn2, was elevated in old cells, contributing to the global reduction in translation without detectable induction of the downstream Gcn4 transcriptional activator. tRNA overexpression activated Gcn2 in young cells and extended lifespan in a manner dependent on Gcn4. Moreover, overexpression of Gcn4 sufficed to extend lifespan in an autophagy-dependent manner in the absence of changes in global translation, indicating that Gcn4-mediated autophagy induction is the ultimate downstream target of activated Gcn2, to extend lifespan.
Collapse
Affiliation(s)
- Zheng Hu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Bo Xia
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States
| | - Spike Dl Postnikoff
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Zih-Jie Shen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Alin S Tomoiaga
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States.,Manhattan College, Bronx, United States
| | - Troy A Harkness
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Ja Hwan Seol
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, United States
| | - Wei Li
- Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States
| | - Kaifu Chen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States
| | - Jessica K Tyler
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| |
Collapse
|
31
|
Detecting senescent fate in mesenchymal stem cells: a combined cytofluorimetric and ultrastructural approach. Biogerontology 2018; 19:401-414. [PMID: 30101381 DOI: 10.1007/s10522-018-9766-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/06/2018] [Indexed: 02/08/2023]
Abstract
Senescence can impair the therapeutic potential of stem cells. In this study, senescence-associated morphofunctional changes in periosteum-derived progenitor cells (PDPCs) from old and young individuals were investigated by combining cytofluorimetry, immunohistochemistry, and transmission electron microscopy. Cell cycle analysis demonstrated a large number of G0/G1 phase cells in PDPCs from old subjects and a progressive accumulation of G0/G1 cells during passaging in cultures from young subjects. Cytofluorimetry documented significant changes in light scattering parameters and closely correlated with the ultrastructural features, especially changes in mitochondrial shape and autophagy, which are consistent with the mitochondrial-lysosomal axis theory of ageing. The combined morphological, biofunctional, and ultrastructural approach enhanced the flow cytometric study of PDPC ageing. We speculate that impaired autophagy, documented in replicative senescent and old PDPCs, reflect a switch from quiescence to senescence. Its demonstration in a tissue with limited turnover-like the cambium layer of the periosteum, where reversible quiescence is the normal stem cell state throughout life-adds a new piece to the regenerative medicine jigsaw in an ageing society.
Collapse
|
32
|
Walters HE, Cox LS. mTORC Inhibitors as Broad-Spectrum Therapeutics for Age-Related Diseases. Int J Mol Sci 2018; 19:E2325. [PMID: 30096787 PMCID: PMC6121351 DOI: 10.3390/ijms19082325] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/22/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
Chronological age represents the greatest risk factor for many life-threatening diseases, including neurodegeneration, cancer, and cardiovascular disease; ageing also increases susceptibility to infectious disease. Current efforts to tackle individual diseases may have little impact on the overall healthspan of older individuals, who would still be vulnerable to other age-related pathologies. However, recent progress in ageing research has highlighted the accumulation of senescent cells with chronological age as a probable underlying cause of pathological ageing. Cellular senescence is an essentially irreversible proliferation arrest mechanism that has important roles in development, wound healing, and preventing cancer, but it may limit tissue function and cause widespread inflammation with age. The serine/threonine kinase mTOR (mechanistic target of rapamycin) is a regulatory nexus that is heavily implicated in both ageing and senescence. Excitingly, a growing body of research has highlighted rapamycin and other mTOR inhibitors as promising treatments for a broad spectrum of age-related pathologies, including neurodegeneration, cancer, immunosenescence, osteoporosis, rheumatoid arthritis, age-related blindness, diabetic nephropathy, muscular dystrophy, and cardiovascular disease. In this review, we assess the use of mTOR inhibitors to treat age-related pathologies, discuss possible molecular mechanisms of action where evidence is available, and consider strategies to minimize undesirable side effects. We also emphasize the urgent need for reliable, non-invasive biomarkers of senescence and biological ageing to better monitor the efficacy of any healthy ageing therapy.
Collapse
Affiliation(s)
- Hannah E Walters
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| | - Lynne S Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
33
|
Dalton HM, Curran SP. Hypodermal responses to protein synthesis inhibition induce systemic developmental arrest and AMPK-dependent survival in Caenorhabditis elegans. PLoS Genet 2018; 14:e1007520. [PMID: 30020921 PMCID: PMC6066256 DOI: 10.1371/journal.pgen.1007520] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/30/2018] [Accepted: 06/27/2018] [Indexed: 01/08/2023] Open
Abstract
Across organisms, manipulation of biosynthetic capacity arrests development early in life, but can increase health- and lifespan post-developmentally. Here we demonstrate that this developmental arrest is not sickness but rather a regulated survival program responding to reduced cellular performance. We inhibited protein synthesis by reducing ribosome biogenesis (rps-11/RPS11 RNAi), translation initiation (ifg-1/EIF3G mutation and egl-45/EIF3A RNAi), or ribosome progression (cycloheximide treatment), all of which result in a specific arrest at larval stage 2 of C. elegans development. This quiescent state can last for weeks—beyond the normal C. elegans adult lifespan—and is reversible, as animals can resume reproduction and live a normal lifespan once released from the source of protein synthesis inhibition. The arrest state affords resistance to thermal, oxidative, and heavy metal stress exposure. In addition to cell-autonomous responses, reducing biosynthetic capacity only in the hypodermis was sufficient to drive organism-level developmental arrest and stress resistance phenotypes. Among the cell non-autonomous responses to protein synthesis inhibition is reduced pharyngeal pumping that is dependent upon AMPK-mediated signaling. The reduced pharyngeal pumping in response to protein synthesis inhibition is recapitulated by exposure to microbes that generate protein synthesis-inhibiting xenobiotics, which may mechanistically reduce ingestion of pathogen and toxin. These data define the existence of a transient arrest-survival state in response to protein synthesis inhibition and provide an evolutionary foundation for the conserved enhancement of healthy aging observed in post-developmental animals with reduced biosynthetic capacity. Protein synthesis is an essential cellular process, but post-developmental reduction of protein synthesis across multiple species leads to improved health- and lifespan. To better understand the physiological responses to impaired protein synthesis, we characterize a novel developmental arrest state that occurs when reducing protein synthesis during C. elegans development. Arrested animals have multiple survival-promoting phenotypes that are all dependent on the cellular energy sensor, AMP kinase. This survival response acts through the hypodermis and causes a reduction in pharyngeal pumping, indicating that the animal is responding to a perceived external threat, even in adults. Furthermore, exposing animals to pathogens, or xenobiotics they produce, can recapitulate these phenotypes, providing a potential evolutionary explanation for how a beneficial response in adults could evolve through the inhibition of an essential biological process such as protein synthesis.
Collapse
Affiliation(s)
- Hans M. Dalton
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
- Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Sean P. Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
- Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California, United States of America
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Nichenametla SN, Mattocks DAL, Malloy VL, Pinto JT. Sulfur amino acid restriction-induced changes in redox-sensitive proteins are associated with slow protein synthesis rates. Ann N Y Acad Sci 2018; 1418:80-94. [PMID: 29377163 DOI: 10.1111/nyas.13556] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/02/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022]
Abstract
The mechanisms underlying life span extension by sulfur amino acid restriction (SAAR) are unclear. Cysteine and methionine are essential for the biosynthesis of proteins and glutathione (GSH), a major redox buffer in the endoplasmic reticulum (ER). We hypothesized that SAAR alters protein synthesis by modulating the redox milieu. Male F344-rats were fed control (CD: 0.86% methionine without cysteine) and SAAR diets (0.17% methionine without cysteine) for 12 weeks. Growth rates, food intake, cysteine and GSH levels, proteins associated with redox status and translation, and fractional protein synthesis rates (FSRs) were determined in liver. Despite a 40% higher food intake, growth rates for SAAR rats were 27% of those fed CD. Hepatic free cysteine in SAAR rats was 55% compared with CD rats. SAAR altered tissue distribution of GSH, as hepatic and erythrocytic levels were 56% and 196% of those in CD rats. Lower GSH levels did not induce ER stress (i.e., unchanged expression of Xbp1s , Chop, and Grp78), but activated PERK and its substrates eIF2-α and NRF2. SAAR-induced changes in translation-initiation machinery (higher p-eIF2-α and 4E-BP1, and lower eIF4G-1) resulted in slower protein synthesis rates (53% of CD). Proteins involved in the antioxidant response (NRF2, KEAP1, GCLM, and NQO1) and protein folding (PDI and ERO1-α) were increased in SAAR. Lower FSR and efficient protein folding might be improving proteostasis in SAAR.
Collapse
Affiliation(s)
| | | | - Virginia L Malloy
- Orentreich Foundation for the Advancement of Science, Cold Spring, New York
| | - John T Pinto
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| |
Collapse
|
35
|
Younis LT, Abu Hassan MI, Taiyeb Ali TB, Bustami TJ. 3D TECA hydrogel reduces cellular senescence and enhances fibroblasts migration in wound healing. Asian J Pharm Sci 2017; 13:317-325. [PMID: 32104405 PMCID: PMC7032142 DOI: 10.1016/j.ajps.2017.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/08/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022] Open
Abstract
This study was designed to investigate the effect of 3D TECA hydrogel on the inflammatory-induced senescence marker, and to assess the influence of the gel on the periodontal ligament fibroblasts (PDLFs) migration in wound healing in vitro. PDLFs were cultured with 20 ng/ml TNF-α to induce inflammation in the presence and absence of 50 µM 3D TECA gel for 14 d. The gel effect on the senescence maker secretory associated-β-galactosidase (SA-β-gal) activity was measured by a histochemical staining. Chromatin condensation and DNA synthesis of the cells were assessed by 4′,6-diamidino-2-phenylindole and 5-ethynyl-2′-deoxyuridine fluorescent staining respectively. For evaluating fibroblasts migration, scratch wound healing assay and Pro-Plus Imaging software were used. The activity of senescence marker, SA-β-gal, was positive in the samples with TNF-α-induced inflammation. SA-β-gal percentage is suppressed (>65%, P < 0.05) in the treated cells with TECA gel as compared to the non-treated cells. Chromatin foci were obvious in the non-treated samples. DNA synthesis was markedly recognized by the fluorescent staining in the treated compared to non-treated cultures. Scratch wound test indicated that the cells migration rate was significantly higher (14.9 µm2/h, P < 0.05) in the treated versus (11 µm2/h) for control PDLFs. The new formula of 3D TECA suppresses the inflammatory-mediated cellular senescence and enhanced fibroblasts proliferation and migration. Therefore, 3D TECA may be used as an adjunct to accelerate repair and healing of periodontal tissues.
Collapse
Affiliation(s)
- Luay Thanoon Younis
- Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh 47000, Malaysia
| | | | - Tara Bai Taiyeb Ali
- Faculty of Dentistry, Universiti Teknologi MARA, MAHSA University, Jenjarom 42610, Malaysia
| | | |
Collapse
|
36
|
Tanaka H, Takebayashi SI, Sakamoto A, Igata T, Nakatsu Y, Saitoh N, Hino S, Nakao M. The SETD8/PR-Set7 Methyltransferase Functions as a Barrier to Prevent Senescence-Associated Metabolic Remodeling. Cell Rep 2017; 18:2148-2161. [PMID: 28249161 DOI: 10.1016/j.celrep.2017.02.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/29/2016] [Accepted: 02/06/2017] [Indexed: 01/01/2023] Open
Abstract
Cellular senescence is an irreversible growth arrest that contributes to development, tumor suppression, and age-related conditions. Senescent cells show active metabolism compared with proliferating cells, but the underlying mechanisms remain unclear. Here we show that the SETD8/PR-Set7 methyltransferase, which catalyzes mono-methylation of histone H4 at lysine 20 (H4K20me1), suppresses nucleolar and mitochondrial activities to prevent cellular senescence. SETD8 protein was selectively downregulated in both oncogene-induced and replicative senescence. Inhibition of SETD8 alone was sufficient to trigger senescence. Under these states, the expression of genes encoding ribosomal proteins (RPs) and ribosomal RNAs as well as the cyclin-dependent kinase (CDK) inhibitor p16INK4A was increased, with a corresponding reduction of H4K20me1 at each locus. As a result, the loss of SETD8 concurrently stimulated nucleolar function and retinoblastoma protein-mediated mitochondrial metabolism. In conclusion, our data demonstrate that SETD8 acts as a barrier to prevent cellular senescence through chromatin-mediated regulation of senescence-associated metabolic remodeling.
Collapse
Affiliation(s)
- Hiroshi Tanaka
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Shin-Ichiro Takebayashi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Akihisa Sakamoto
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Tomoka Igata
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yuko Nakatsu
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Noriko Saitoh
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan.
| |
Collapse
|
37
|
Nacarelli T, Sell C. Targeting metabolism in cellular senescence, a role for intervention. Mol Cell Endocrinol 2017; 455:83-92. [PMID: 27591812 DOI: 10.1016/j.mce.2016.08.049] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 07/29/2016] [Accepted: 08/30/2016] [Indexed: 01/06/2023]
Abstract
Cellular senescence has gained much attention as a contributor to aging and susceptibility to disease. Senescent cells undergo a stable cell cycle arrest and produce pro-inflammatory cytokines. However, an additional feature of the senescence phenotype is an altered metabolic state. Despite maintaining a non-dividing state, senescent cells display a high metabolic rate. Metabolic changes characteristic of replicative senescence include altered mitochondrial function and perturbations in growth signaling pathways, such as the mTORC1-signaling pathway. Recent evidence has raised the possibility that these metabolic changes may be essential for the induction and maintenance of the senescent state. Interventions such as rapamycin treatment and methionine restriction impact key aspects of metabolism and delay cellular senescence to extend cellular lifespan. Here, we review the metabolic changes and potential metabolic regulators of the senescence program. In addition, we will discuss how lifespan-extending regimens prevent metabolic stress that accompanies and potentially regulates the senescence program.
Collapse
Affiliation(s)
- Timothy Nacarelli
- Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Christian Sell
- Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| |
Collapse
|
38
|
Saneja A, Nayak D, Srinivas M, Kumar A, Khare V, Katoch A, Goswami A, Vishwakarma RA, Sawant SD, Gupta PN. Development and mechanistic insight into enhanced cytotoxic potential of hyaluronic acid conjugated nanoparticles in CD44 overexpressing cancer cells. Eur J Pharm Sci 2016; 97:79-91. [PMID: 27989859 DOI: 10.1016/j.ejps.2016.10.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/03/2016] [Accepted: 10/27/2016] [Indexed: 01/08/2023]
Abstract
The overexpression of CD44 in cancer cells reroutes number of oncogenic pathways including the central Pi3K/Akt/NF-kB pathway leading to cancer progression and malignancy. Herein, we developed hyaluronic acid-modified poly(dl-lactic-co-glycolic acid)-poly (ethylene glycol) nanoparticles (PLGA-PEG-HA NPs) for targeted delivery of TTQ (thio-tetrazolyl analog of a clinical candidate, IC87114) to CD44 overexpressing cancer cells. The PLGA-PEG co-polymer was synthesized and characterized by NMR and FTIR. The co-polymer based nanoparticles were prepared by solvent evaporation method and hyaluronic acid (HA) was conjugated on to the nanoparticle surface via EDC/NHS chemistry. The PLGA-PEG-HA NPs had a desirable particle size (<200nm) with reduced polydispersibility and exhibited spherical shape under atomic force microscope (AFM). In vitro cytotoxicity and cellular uptake studies demonstrated higher cytotoxicity and enhanced intracellular accumulation of PLGA-PEG-HA NPs compared to PLGA-PEG NPs in high CD44 expressing MiaPaca-2 cells compared to MDA-MB-231 and MCF7 cells. At the molecular level, the PLGA-PEG-HA NPs were found to be inducing premature senescence with increase in senescence associated β-galactosidase activity and senescence specific marker p21 expression through modulation of Pi3K/Akt/NF-kB signaling pathway in MiaPaca-2 cells. These findings collectively indicated that HA-modified nanoparticles might serve as a promising nanocarrier for site-specific drug delivery, and can be explored further to increase the therapeutic efficacy of anticancer drugs via targeting to CD44 over-expressing cancer cells.
Collapse
Affiliation(s)
- Ankit Saneja
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Debasis Nayak
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - M Srinivas
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Amit Kumar
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Vaibhav Khare
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Archana Katoch
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Anindya Goswami
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Ram A Vishwakarma
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Sanghapal D Sawant
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Prem N Gupta
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India; Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| |
Collapse
|
39
|
Takauji Y, Miki K, Mita J, Hossain MN, Yamauchi M, Kioi M, Ayusawa D, Fujii M. Triphala, a formulation of traditional Ayurvedic medicine, shows protective effect against X-radiation in HeLa cells. J Biosci 2016; 41:569-575. [DOI: 10.1007/s12038-016-9639-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
40
|
Kudo I, Nozawa M, Miki K, Takauji Y, En A, Fujii M, Ayusawa D. Dual roles of ERK1/2 in cellular senescence induced by excess thymidine in HeLa cells. Exp Cell Res 2016; 346:216-23. [PMID: 27443255 DOI: 10.1016/j.yexcr.2016.07.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/11/2016] [Accepted: 07/17/2016] [Indexed: 12/23/2022]
Abstract
DNA damage response is crucially involved in cellular senescence. We have previously shown that excess thymidine, which stalls DNA replication forks, induces cellular senescence in human cells, and ERK1/2 play a key role in the induction of it. In this study, we found that Chk1 and ERK1/2 were activated to promote cell survival upon addition of excess thymidine. Knockdown of ERK1/2 activated Chk1, and conversely, knockdown of Chk1 activated ERK1/2, which observations suggested a mechanism for compensatory activation of Chk1 and ERK1/2 in the absence of ERK1/2 and Chk1, respectively. We also found that Chk1 functioned mainly at the onset of cellular senescence, and on the other hand, ERK1/2 functioned for a more extended period to induce cellular senescence. Our findings suggested that Chk1 and ERK1/2 were activated to promote cell survival upon addition of excess thymidine, but prolonged activation of ERK1/2 led to cellular senescence. This implies a pleiotropic effect of ERK1/2 in cellular senescence induced by excess thymidine.
Collapse
Affiliation(s)
- Ikuru Kudo
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Megumi Nozawa
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Kensuke Miki
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| | - Yuki Takauji
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Atsuki En
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Michihiko Fujii
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan.
| | - Dai Ayusawa
- Graduate school of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| |
Collapse
|
41
|
Takauji Y, En A, Miki K, Ayusawa D, Fujii M. Combinatorial effects of continuous protein synthesis, ERK-signaling, and reactive oxygen species on induction of cellular senescence. Exp Cell Res 2016; 345:239-46. [PMID: 27339653 DOI: 10.1016/j.yexcr.2016.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/14/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022]
Abstract
Mammalian cells, when treated with sub-lethal doses of genotoxic stresses, slow down DNA synthesis but continue protein synthesis. Thus, these cells show an accumulation of proteins and undergo unbalanced growth. In the previous studies, we have shown that HeLa cells treated with excess thymidine or camptothecin undergo unbalanced growth, and prolonged unbalanced growth causes induction of cellular senescence, which is suppressed by restriction of protein synthesis or inhibition of ERK-signaling. In this study, we found that restriction of protein synthesis, inhibition of ERK-signaling, and elimination of reactive oxygen species showed a combinatorial effect on suppression of cellular senescence induced by excess thymidine or camptothecin. Of these, restriction of protein synthesis most effectively suppressed cellular senescence. Importantly, a similar combinatorial effect was observed in replicative senescence in normal human diploid fibroblasts. Our findings suggested that various stresses were cumulatively involved in cellular senescence, and suppression of cellular senescence was improved by combining the treatments that reduce the stresses.
Collapse
Affiliation(s)
- Yuki Takauji
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Atsuki En
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Kensuke Miki
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| | - Dai Ayusawa
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan; Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| | - Michihiko Fujii
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan.
| |
Collapse
|