1
|
Liang R, Qi X, Cai Q, Niu L, Huang X, Zhang D, Ling J, Wu Y, Chen Y, Yang P, Liu J, Zhang J, Yu P. The role of NLRP3 inflammasome in aging and age-related diseases. Immun Ageing 2024; 21:14. [PMID: 38317229 PMCID: PMC10840156 DOI: 10.1186/s12979-023-00395-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 02/07/2024]
Abstract
The gradual aging of the global population has led to a surge in age-related diseases, which seriously threaten human health. Researchers are dedicated to understanding and coping with the complexities of aging, constantly uncovering the substances and mechanism related to aging like chronic low-grade inflammation. The NOD-like receptor protein 3 (NLRP3), a key regulator of the innate immune response, recognizes molecular patterns associated with pathogens and injury, initiating an intrinsic inflammatory immune response. Dysfunctional NLRP3 is linked to the onset of related diseases, particularly in the context of aging. Therefore, a profound comprehension of the regulatory mechanisms of the NLRP3 inflammasome in aging-related diseases holds the potential to enhance treatment strategies for these conditions. In this article, we review the significance of the NLRP3 inflammasome in the initiation and progression of diverse aging-related diseases. Furthermore, we explore preventive and therapeutic strategies for aging and related diseases by manipulating the NLRP3 inflammasome, along with its upstream and downstream mechanisms.
Collapse
Affiliation(s)
- Ruikai Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Xinrui Qi
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Qi Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Liyan Niu
- Huan Kui College of Nanchang University, Nanchang, China
| | - Xi Huang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China.
| |
Collapse
|
2
|
Metformin Attenuates Inflammation and Fibrosis in Thyroid-Associated Ophthalmopathy. Int J Mol Sci 2022; 23:ijms232415508. [PMID: 36555150 PMCID: PMC9778898 DOI: 10.3390/ijms232415508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of thyroid-associated ophthalmopathy (TAO) is still unclear, and therapeutic drugs have great limitations. As metformin has multiple therapeutic effects in many autoimmune diseases, we explored the effects of metformin on TAO in an in vitro fibroblast model. We used orbital connective tissues and fibroblasts that were obtained from TAO patients and normal controls. The activity of adenosine monophosphate-activated protein kinase (AMPK) and the levels of inflammatory or fibrotic factors were examined by immunofluorescence (IF) and immunohistochemistry (IHC). Quantitative real-time polymerase chain reaction (qPCR), cytokine quantification by enzyme-linked immunosorbent sssay (ELISA), IF, and western blotting (WB) were used to measure the expression of factors related to inflammation, fibrosis, and autophagy. To determine the anti-inflammatory and antifibrotic mechanisms of metformin, we pretreated cells with metformin, 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR, an AMPK activator) or compound C (CC, an AMPK inhibitor) for 24 h and used WB to verify the changes in protein levels in the AMPK/mammalian target of rapamycin (mTOR) pathway. We determined that the low activity of AMPK in the periorbital tissue of TAO patients may be closely related to the occurrence and development of inflammation and fibrosis, and metformin exerts multiple effects by activating AMPK in TAO. Furthermore, we suggest that AMPK may be a potential target of TAO therapy.
Collapse
|
3
|
Espinoza SE, Woods RL, Ekram ARMS, Ernst ME, Polekhina G, Wolfe R, Shah RC, Ward SA, Storey E, Nelson MR, Reid CM, Lockery JE, Orchard SG, Trevaks R, Fitzgerald SM, Stocks NP, Chan A, McNeil JJ, Murray AM, Newman AB, Ryan J. The Effect of Low-Dose Aspirin on Frailty Phenotype and Frailty Index in Community-Dwelling Older Adults in the ASPirin in Reducing Events in the Elderly Study. J Gerontol A Biol Sci Med Sci 2022; 77:2007-2014. [PMID: 34758073 PMCID: PMC9536436 DOI: 10.1093/gerona/glab340] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Frailty is associated with chronic inflammation, which may be modified by aspirin. The purpose of this study was to determine whether low-dose aspirin reduces incident frailty in healthy older adult participants of the ASPirin in Reducing Events in the Elderly (ASPREE) trial. METHODS In the United States and Australia, 19 114 community-dwelling individuals aged ≥70 and older (U.S. minorities ≥65 years) and free of overt cardiovascular disease, persistent physical disability, and dementia were enrolled in ASPREE, a double-blind, placebo-controlled trial of 100-mg daily aspirin versus placebo. Frailty, a prespecified study end point, was defined according to a modified Fried frailty definition (Fried frailty) and the frailty index based on the deficit accumulation model (frailty index). Competing risk Cox proportional hazard models were used to compare time to incident frailty by aspirin versus placebo. Sensitivity analysis was conducted to include frailty data with and without imputation of missing data. RESULTS Over a median 4.7 years, 2 252 participants developed incident Fried frailty, and 4 451 had incident frailty according to the frailty index. Compared with placebo, aspirin treatment did not alter the risk of incident frailty (Fried frailty hazard ratio [HR]: 1.04, 95% confidence interval [CI] 0.96-1.13; frailty index HR: 1.03, 95% CI 0.97-1.09). The proportion of individuals classified as frail, and the trajectory in continuous frailty scores over time, were not different between the aspirin and placebo treatment groups. The results were consistent across a series of subgroups. CONCLUSIONS Low-dose aspirin use in healthy older adults when initiated in older ages does not reduce risk of incident frailty or the trajectory of frailty.
Collapse
Affiliation(s)
- Sara E Espinoza
- Division of Geriatrics, Gerontology and Palliative Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA.,Geriatrics Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - A R M Saifuddin Ekram
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Iowa, Iowa City, Iowa, USA.,Department of Family Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Galina Polekhina
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Raj C Shah
- Department of Family Medicine and Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Stephanie A Ward
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elsdon Storey
- Van Cleef/Roet Centre for Nervous Diseases, Monash University, Melbourne, Victoria, Australia
| | - Mark R Nelson
- Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | - Christopher M Reid
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jessica E Lockery
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Suzanne G Orchard
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ruth Trevaks
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sharyn M Fitzgerald
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nigel P Stocks
- Discipline of General Practice, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Andy Chan
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John J McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anne M Murray
- Berman Center for Clinical Outcomes and Research, Hennepin Health Research Institute and Division of Geriatrics, Department of Medicine, Hennepin Healthcare and University of Minnesota, Minneapolis, Minnesota, USA
| | - Anne B Newman
- Center for Aging and Population Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joanne Ryan
- Geriatrics Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | | |
Collapse
|
4
|
Oxidative stress, aging, antioxidant supplementation and their impact on human health: An overview. Mech Ageing Dev 2022; 206:111707. [PMID: 35839856 DOI: 10.1016/j.mad.2022.111707] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/06/2022] [Accepted: 07/10/2022] [Indexed: 12/12/2022]
Abstract
Aging is characterized by a progressive loss of tissue and organ function due to genetic and environmental factors, nutrition, and lifestyle. Oxidative stress is one the most important mechanisms of cellular senescence and increased frailty, resulting in several age-linked, noncommunicable diseases. Contributing events include genomic instability, telomere shortening, epigenetic mechanisms, reduced proteome homeostasis, altered stem-cell function, defective intercellular communication, progressive deregulation of nutrient sensing, mitochondrial dysfunction, and metabolic unbalance. These complex events and their interplay can be modulated by dietary habits and the ageing process, acting as potential measures of primary and secondary prevention. Promising nutritional approaches include the Mediterranean diet, the intake of dietary antioxidants, and the restriction of caloric intake. A comprehensive understanding of the ageing processes should promote new biomarkers of risk or diagnosis, but also beneficial treatments oriented to increase lifespan.
Collapse
|
5
|
Fischer F, Grigolon G, Benner C, Ristow M. Evolutionarily conserved transcription factors as regulators of longevity and targets for geroprotection. Physiol Rev 2022; 102:1449-1494. [PMID: 35343830 DOI: 10.1152/physrev.00017.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aging is the single largest risk factor for many debilitating conditions, including heart diseases, stroke, cancer, diabetes, and neurodegenerative disorders. While far from understood in its full complexity, it is scientifically well-established that aging is influenced by genetic and environmental factors, and can be modulated by various interventions. One of aging's early hallmarks are aberrations in transcriptional networks, controlling for example metabolic homeostasis or the response to stress. Evidence in different model organisms abounds that a number of evolutionarily conserved transcription factors, which control such networks, can affect lifespan and healthspan across species. These transcription factors thus potentially represent conserved regulators of longevity and are emerging as important targets in the challenging quest to develop treatments to mitigate age-related diseases, and possibly even to slow aging itself. This review provides an overview of evolutionarily conserved transcription factors that impact longevity or age-related diseases in at least one multicellular model organism (nematodes, flies, or mice), and/or are tentatively linked to human aging. Discussed is the general evidence for transcriptional regulation of aging and disease, followed by a more detailed look at selected transcription factor families, the common metabolic pathways involved, and the targeting of transcription factors as a strategy for geroprotective interventions.
Collapse
Affiliation(s)
- Fabian Fischer
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Giovanna Grigolon
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Christoph Benner
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
6
|
Restoration of aged hematopoietic cells by their young counterparts through instructive microvesicles release. Aging (Albany NY) 2021; 13:23981-24016. [PMID: 34762598 PMCID: PMC8610119 DOI: 10.18632/aging.203689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022]
Abstract
This study addresses the potential to reverse age-associated morbidity by establishing methods to restore the aged hematopoietic system. Parabiotic animal models indicated that young secretome could restore aged tissues, leading us to establish a heterochronic transwell system with aged mobilized peripheral blood (MPB), co-cultured with young MPB or umbilical cord blood (UCB) cells. Functional studies and omics approaches indicate that the miRNA cargo of microvesicles (MVs) restores the aged hematopoietic system. The in vitro findings were validated in immune deficient (NSG) mice carrying an aged hematopoietic system, improving aged hallmarks such as increased lymphoid:myeloid ratio, decreased inflammation and cellular senescence. Elevated MYC and E2F pathways, and decreased p53 were key to hematopoietic restoration. These processes require four restorative miRs that target the genes for transcription/differentiation, namely PAX and phosphatase PPMIF. These miRs when introduced in aged cells were sufficient to restore the aged hematopoietic system in NSG mice. The aged MPBs were the drivers of their own restoration, as evidenced by the changes from distinct baseline miR profiles in MPBs and UCB to comparable expressions after exposure to aged MPBs. Restorative natural killer cells eliminated dormant breast cancer cells in vivo, indicating the broad relevance of this cellular paradigm - preventing and reversing age-associated disorders such as clearance of early malignancies and enhanced responses to vaccine and infection.
Collapse
|
7
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kamperi N, Kanara I, Kodukula K, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Steliou K, Tamvakopoulos C, Vavvas DG, Zamboni RJ, Sampani K. Pathogenic mitochondrial dysfunction and metabolic abnormalities. Biochem Pharmacol 2021; 193:114809. [PMID: 34673016 DOI: 10.1016/j.bcp.2021.114809] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Abstract
Herein we trace links between biochemical pathways, pathogenesis, and metabolic diseases to set the stage for new therapeutic advances. Cellular and acellular microorganisms including bacteria and viruses are primary pathogenic drivers that cause disease. Missing from this statement are subcellular compartments, importantly mitochondria, which can be pathogenic by themselves, also serving as key metabolic disease intermediaries. The breakdown of food molecules provides chemical energy to power cellular processes, with mitochondria as powerhouses and ATP as the principal energy carrying molecule. Most animal cell ATP is produced by mitochondrial synthase; its central role in metabolism has been known for >80 years. Metabolic disorders involving many organ systems are prevalent in all age groups. Progressive pathogenic mitochondrial dysfunction is a hallmark of genetic mitochondrial diseases, the most common phenotypic expression of inherited metabolic disorders. Confluent genetic, metabolic, and mitochondrial axes surface in diabetes, heart failure, neurodegenerative disease, and even in the ongoing coronavirus pandemic.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, NY, USA
| | - David N Harpp
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Natalia Kamperi
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | | | - Anastasios N Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Whitney R Powers
- Department of Health Sciences, Boston University, Boston, MA, USA; Department of Anatomy, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA; PhenoMatriX, Inc., Natick, MA, USA
| | - Constantin Tamvakopoulos
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Demetrios G Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Robert J Zamboni
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Konstantina Sampani
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Statzer C, Jongsma E, Liu SX, Dakhovnik A, Wandrey F, Mozharovskyi P, Zülli F, Ewald CY. Youthful and age-related matreotypes predict drugs promoting longevity. Aging Cell 2021; 20:e13441. [PMID: 34346557 PMCID: PMC8441316 DOI: 10.1111/acel.13441] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/16/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
The identification and validation of drugs that promote health during aging ("geroprotectors") are key to the retardation or prevention of chronic age-related diseases. Here, we found that most of the established pro-longevity compounds shown to extend lifespan in model organisms also alter extracellular matrix gene expression (i.e., matrisome) in human cell lines. To harness this observation, we used age-stratified human transcriptomes to define the age-related matreotype, which represents the matrisome gene expression pattern associated with age. Using a "youthful" matreotype, we screened in silico for geroprotective drug candidates. To validate drug candidates, we developed a novel tool using prolonged collagen expression as a non-invasive and in-vivo surrogate marker for Caenorhabditis elegans longevity. With this reporter, we were able to eliminate false-positive drug candidates and determine the appropriate dose for extending the lifespan of C. elegans. We improved drug uptake for one of our predicted compounds, genistein, and reconciled previous contradictory reports of its effects on longevity. We identified and validated new compounds, tretinoin, chondroitin sulfate, and hyaluronic acid, for their ability to restore age-related decline of collagen homeostasis and increase lifespan. Thus, our innovative drug screening approach-employing extracellular matrix homeostasis-facilitates the discovery of pharmacological interventions promoting healthy aging.
Collapse
Affiliation(s)
- Cyril Statzer
- Department of Health Sciences and TechnologyInstitute of Translational MedicineEidgenössische Technische Hochschule ZürichSchwerzenbach‐ZürichSwitzerland
| | - Elisabeth Jongsma
- Department of Health Sciences and TechnologyInstitute of Translational MedicineEidgenössische Technische Hochschule ZürichSchwerzenbach‐ZürichSwitzerland
| | - Sean X. Liu
- Department of Health Sciences and TechnologyInstitute of Translational MedicineEidgenössische Technische Hochschule ZürichSchwerzenbach‐ZürichSwitzerland
| | - Alexander Dakhovnik
- Department of Health Sciences and TechnologyInstitute of Translational MedicineEidgenössische Technische Hochschule ZürichSchwerzenbach‐ZürichSwitzerland
| | | | | | - Fred Zülli
- Mibelle Biochemistry, Mibelle AGBuchsSwitzerland
| | - Collin Y. Ewald
- Department of Health Sciences and TechnologyInstitute of Translational MedicineEidgenössische Technische Hochschule ZürichSchwerzenbach‐ZürichSwitzerland
| |
Collapse
|
9
|
Vertti-Quintero N, Berger S, Casadevall I Solvas X, Statzer C, Annis J, Ruppen P, Stavrakis S, Ewald CY, Gunawan R, deMello AJ. Stochastic and Age-Dependent Proteostasis Decline Underlies Heterogeneity in Heat-Shock Response Dynamics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102145. [PMID: 34196492 DOI: 10.1002/smll.202102145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Indexed: 06/13/2023]
Abstract
Significant non-genetic stochastic factors affect aging, causing lifespan differences among individuals, even those sharing the same genetic and environmental background. In Caenorhabditis elegans, differences in heat-shock response (HSR) are predictive of lifespan. However, factors contributing to the heterogeneity of HSR are still not fully elucidated. Here, the authors characterized HSR dynamics in isogenic C. elegans expressing GFP reporter for hsp-16.2 for identifying the key contributors of HSR heterogeneity. Specifically, microfluidic devices that enable cross-sectional and longitudinal measurements of HSR dynamics in C. elegans at different scales are developed: in populations, within individuals, and in embryos. The authors adapted a mathematical model of HSR to single C. elegans and identified model parameters associated with proteostasis-maintenance of protein homeostasis-more specifically, protein turnover, as the major drivers of heterogeneity in HSR dynamics. It is verified that individuals with enhanced proteostasis fidelity in early adulthood live longer. The model-based comparative analysis of protein turnover in day-1 and day-2 adult C. elegans revealed a stochastic-onset of age-related proteostasis decline that increases the heterogeneity of HSR capacity. Finally, the analysis of C. elegans embryos showed higher HSR and proteostasis capacity than young adults and established transgenerational contribution to HSR heterogeneity that depends on maternal age.
Collapse
Affiliation(s)
| | - Simon Berger
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Xavier Casadevall I Solvas
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
- Department of Biosystems, KU Leuven, Leuven, B-3001, Belgium
| | - Cyril Statzer
- Institute of Translational Medicine, ETH Zurich, Schwerzenbach, 8603, Switzerland
| | - Jillian Annis
- Department of Chemical and Biological Engineering, University at Buffalo - SUNY, Buffalo, NY, 14260, USA
| | - Peter Ruppen
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Stavros Stavrakis
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Collin Y Ewald
- Institute of Translational Medicine, ETH Zurich, Schwerzenbach, 8603, Switzerland
| | - Rudiyanto Gunawan
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
- Department of Chemical and Biological Engineering, University at Buffalo - SUNY, Buffalo, NY, 14260, USA
| | - Andrew J deMello
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| |
Collapse
|
10
|
Cereblon contributes to the development of pulmonary fibrosis via inactivation of adenosine monophosphate-activated protein kinase α1. Exp Mol Med 2021; 53:885-893. [PMID: 34002012 PMCID: PMC8178361 DOI: 10.1038/s12276-021-00619-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 11/09/2022] Open
Abstract
Pulmonary fibrosis is a progressive and lethal lung disease characterized by the proliferation and differentiation of lung fibroblasts and the accumulation of extracellular matrices. Since pulmonary fibrosis was reported to be associated with adenosine monophosphate-activated protein kinase (AMPK) activation, which is negatively regulated by cereblon (CRBN), we aimed to determine whether CRBN is involved in the development of pulmonary fibrosis. Therefore, we evaluated the role of CRBN in bleomycin (BLM)-induced pulmonary fibrosis in mice and in transforming growth factor-beta 1 (TGF-β1)-induced differentiation of human lung fibroblasts. BLM-induced fibrosis and the mRNA expression of collagen and fibronectin were increased in the lung tissues of wild-type (WT) mice; however, they were significantly suppressed in Crbn knockout (KO) mice. While the concentrations of TGF-β1/2 in bronchoalveolar lavage fluid were increased via BLM treatment, they were similar between BLM-treated WT and Crbn KO mice. Knockdown of CRBN suppressed TGF-β1-induced activation of small mothers against decapentaplegic 3 (SMAD3), and overexpression of CRBN increased it. TGF-β1-induced activation of SMAD3 increased α-smooth muscle actin (α-SMA) and collagen levels. CRBN was found to be colocalized with AMPKα1 in lung fibroblasts. CRBN overexpression inactivated AMPKα1. When cells were treated with metformin (an AMPK activator), the CRBN-induced activation of SMAD3 and upregulation of α-SMA and collagen expression were significantly suppressed, suggesting that increased TGF-β1-induced activation of SMAD3 via CRBN overexpression is associated with AMPKα1 inactivation. Taken together, these data suggest that CRBN is a profibrotic regulator and maybe a potential target for treating lung fibrosis. Interventions that target a regulatory protein called cereblon could help reduce the damage inflicted on the lungs by idiopathic pulmonary fibrosis (IPF). This incurable and generally fatal condition is associated with the accumulation of scar tissue in the lungs, which leads to the gradual loss of respiratory function. Researchers led by Kyoung-Hee Lee at Seoul National University Hospital in South Korea have now identified cereblon as a potentially important contributor to this scarring process. They found that cereblon regulates a complex metabolic pathway that ultimately contributes to production of fibrosis-related proteins in a mouse model of IPF. Genetically modified animals that lacked the gene encoding cereblon showed reduced accumulation of these proteins in their lungs. These results suggest that cereblon-inhibiting agents could potentially control the progression of IPF and help preserve lung function.
Collapse
|
11
|
Syal C, Kosaraju J, Hamilton L, Aumont A, Chu A, Sarma SN, Thomas J, Seegobin M, Dilworth FJ, He L, Wondisford FE, Zimmermann R, Parent M, Fernandes K, Wang J. Dysregulated expression of monoacylglycerol lipase is a marker for anti-diabetic drug metformin-targeted therapy to correct impaired neurogenesis and spatial memory in Alzheimer's disease. Am J Cancer Res 2020; 10:6337-6360. [PMID: 32483456 PMCID: PMC7255032 DOI: 10.7150/thno.44962] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: Monoacylglycerol lipase (Mgll), a hydrolase that breaks down the endocannabinoid 2-arachidonoyl glycerol (2-AG) to produce arachidonic acid (ARA), is a potential target for neurodegenerative diseases, such as Alzheimer's disease (AD). Increasing evidence shows that impairment of adult neurogenesis by perturbed lipid metabolism predisposes patients to AD. However, it remains unknown what causes aberrant expression of Mgll in AD and how Mgll-regulated lipid metabolism impacts adult neurogenesis, thus predisposing to AD during aging. Here, we identify Mgll as an aging-induced factor that impairs adult neurogenesis and spatial memory in AD, and show that metformin, an FDA-approved anti-diabetic drug, can reduce the expression of Mgll to reverse impaired adult neurogenesis, prevent spatial memory decline and reduce β-amyloid accumulation. Methods: Mgll expression was assessed in both human AD patient post-mortem hippocampal tissues and 3xTg-AD mouse model. In addition, we used both the 3xTg-AD animal model and the CbpS436A genetic knock-in mouse model to identify that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway, involving atypical protein kinase C (aPKC)-stimulated Ser436 phosphorylation of histone acetyltransferase CBP through biochemical methods. Furthermore, we performed in vivo adult neurogenesis assay with BrdU/EdU labelling and Morris water maze task in both animal models following pharmacological treatments to show the key role of Mgll in metformin-corrected neurogenesis and spatial memory deficits of AD through reactivating the aPKC-CBP pathway. Finally, we performed in vitro adult neurosphere assays using both animal models to study the role of the aPKC-CBP mediated Mgll repression in determining adult neural stem/progenitor cell (NPC) fate. Results: Here, we demonstrate that aging-dependent induction of Mgll is observed in the 3xTg-AD model and human AD patient post-mortem hippocampal tissues. Importantly, we discover that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway. The accumulation of Mgll in the 3xTg-AD mice reduces the genesis of newborn neurons and perturbs spatial memory. However, we find that metformin-stimulated aPKC-CBP pathway decreases Mgll expression to recover these deficits in 3xTg-AD. In addition, we reveal that elevated Mgll levels in cultured adult NPCs from both 3xTg-AD and CbpS436A animal models are responsible for their NPC neuronal differentiation deficits. Conclusion: Our findings set the stage for development of a clinical protocol where Mgll would serve as a biomarker in early stages of AD to identify potential metformin-responsive AD patients to restore their neurogenesis and spatial memory.
Collapse
|
12
|
Zhang S, Li F, Zhou T, Wang G, Li Z. Caenorhabditis elegans as a Useful Model for Studying Aging Mutations. Front Endocrinol (Lausanne) 2020; 11:554994. [PMID: 33123086 PMCID: PMC7570440 DOI: 10.3389/fendo.2020.554994] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
The Caenorhabditis elegans genome possesses homologs of about two-thirds of all human disease genes. Based on its physiological aging characteristics and superiority, the use of C. elegans as a model system for studies on aging, age-related diseases, mechanisms of longevity, and drug screening has been widely acknowledged in recent decades. Lifespan increasing mutations in C. elegans were found to delay aging by impinging several signaling pathways and related epigenetic modifications, including the insulin/IGF-1 signaling (IIS), AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR) pathways. Interestingly, dietary restriction (DR) has been shown to increase the lifespan of numerous metazoans and protect them from multiple age-related pathologies. However, the underlying molecular mechanisms are unclear. In recent decades, C. elegans has been used as a unique model system for high-throughput drug screening. Here, we review C. elegans mutants exhibiting increased in lifespan and age-dependent changes under DR, as well as the utility of C. elegans for drug screening. Thus, we provide evidence for the use of this model organism in research on the prevention of aging.
Collapse
|
13
|
Fulop T, Larbi A, Khalil A, Cohen AA, Witkowski JM. Are We Ill Because We Age? Front Physiol 2019; 10:1508. [PMID: 31956310 PMCID: PMC6951428 DOI: 10.3389/fphys.2019.01508] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
Growing elderly populations, sometimes referred to as gray (or silver) tsunami, are an increasingly serious health and socioeconomic concern for modern societies. Science has made tremendous progress in the understanding of aging itself, which has helped medicine to extend life expectancies. With the increase of the life expectancy, the incidence of chronic age-related diseases (ARDs) has also increased. A new approach trying to solve this problem is the concept of geroscience. This concept implies that the aging process itself is the common cause of all ARDs. The corollary and consequence of such thinking is that we can and should treat aging itself as a disease. How to translate this into the medical practice is a big challenge, but if we consider aging as a disease the problem is solved. However, as there is no common definition of what aging is, what its causes are, why it occurs, and what should be the target(s) for interventions, it is impossible to conclude that aging is a disease. On the contrary, aging should be strongly considered not to be a disease and as such should not be treated; nonetheless, aging is likely amenable to optimization of changes/adaptations at an individual level to achieve a better functional healthspan.
Collapse
Affiliation(s)
- Tamas Fulop
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Biopolis, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, University of Singapore, Singapore, Singapore
- Department of Biology, Faculty of Sciences, University of Tunis El Manar, Tunis, Tunisia
| | - Abdelouahed Khalil
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Alan A. Cohen
- Department of Family Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
14
|
Mattedi F, Vagnoni A. Temporal Control of Axonal Transport: The Extreme Case of Organismal Ageing. Front Cell Neurosci 2019; 13:393. [PMID: 31555095 PMCID: PMC6716446 DOI: 10.3389/fncel.2019.00393] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/09/2019] [Indexed: 01/05/2023] Open
Abstract
A fundamental question in cell biology is how cellular components are delivered to their destination with spatial and temporal precision within the crowded cytoplasmic environment. The long processes of neurons represent a significant spatial challenge and make these cells particularly dependent on mechanisms for long-range cytoskeletal transport of proteins, RNA and organelles. Although many studies have substantiated a role for defective transport of axonal cargoes in the pathogenesis of neurodevelopmental and neurodegenerative diseases, remarkably little is known about how transport is regulated throughout ageing. The scale of the challenge posed by ageing is considerable because, in this case, the temporal regulation of transport is ultimately dictated by the length of organismal lifespan, which can extend to days, years or decades. Recent methodological advances to study live axonal transport during ageing in situ have provided new tools to scratch beneath the surface of this complex problem and revealed that age-dependent decline in the transport of mitochondria is a common feature across different neuronal populations of several model organisms. In certain instances, the molecular pathways that affect transport in ageing animals have begun to emerge. However, the functional implications of these observations are still not fully understood. Whether transport decline is a significant determinant of neuronal ageing or a mere consequence of decreased cellular fitness remains an open question. In this review, we discuss the latest developments in axonal trafficking in the ageing nervous system, along with the early studies that inaugurated this new area of research. We explore the possibility that the interplay between mitochondrial function and motility represents a crucial driver of ageing in neurons and put forward the hypothesis that declining axonal transport may be legitimately considered a hallmark of neuronal ageing.
Collapse
Affiliation(s)
| | - Alessio Vagnoni
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, IoPPN, King’s College London, London, United Kingdom
| |
Collapse
|
15
|
Gao J, Cui JZ, Wang A, Chen HHR, Fong A, Matsubara JA. The reduction of XIAP is associated with inflammasome activation in RPE: implications for AMD pathogenesis. J Neuroinflammation 2019; 16:171. [PMID: 31438981 PMCID: PMC6706877 DOI: 10.1186/s12974-019-1558-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Background Age-related macular degeneration (AMD) is a multifactorial chronic disease of the eye. Several candidate pathways have been hypothesized to play a role in AMD pathogenesis. Our work and those of others suggests inflammasome activity as a mechanism associated with retinal pigment epithelial (RPE) cell demise. X-linked inhibitor of apoptosis protein (XIAP), an anti-apoptosis factor, has recently been shown to regulate inflammasome activity in non-ocular cells. The purpose of this study is to characterize XIAP’s regulatory role in RPE. Methods Protein lysates of eye tissues from rats (vinpocetine- or aurin tricarboxylic acid complex-treated, ATAC, vs naïve) and mice (wild type vs Caspase-4−/−) were utilized to analyze XIAP protein levels. Immunohistochemistry was used to detect NLRP3 levels in the RPE layer. In vitro inflammasome activation on RPE cells was achieved with L-leucyl-L-leucine methyl ester (Leu-Leu-OMe) stimulation. Levels of XIAP mRNA and 18S RNA were quantified by RT-PCR. Cell culture supernatants were tested directly for secreted IL-1β by ELISA or concentrated for the detection of secreted IL-18 by western blot. Protein lysates from RPE in cell culture were collected for the measurement of cleaved caspase-1 p20, XIAP, and GAPDH. Data are presented as Mean ± SD. p < 0.05 is considered statistically significant. Results The XIAP protein level was significantly increased when the inflammasome was inhibited at the “activation” step by ATAC, but not the “priming” step, in vivo. Concomitantly, NLRP3 immunoreactivity was lower in the RPE layer of animals fed with ATAC. In mice where caspase-1 cleavage was impaired by the genetic deficiency in caspase-4, the XIAP protein level increased in eye tissues. In RPE cell culture, Leu-Leu-OMe stimulation led to caspase-1 cleavage, cytokine secretion, and XIAP reduction, which can be abolished by Z-YVAD-FMK. When XIAP siRNA was given as a pre-treatment to RPE in vitro, Leu-Leu-OMe induced IL-1β/IL-18 secretion was enhanced, whereas overexpressing XIAP reduced IL-1β secretion under inflammasome activation, both compared to controls cells. Conclusions Together, these data suggest XIAP-mediated inhibition of inflammasome activity in RPE may provide insights into the biological consequences of inflammasome activation in RPE and reveals the caspase-1/XIAP/IL-1β/IL-18 axis as a target for broader applications in AMD biology and treatment design. Electronic supplementary material The online version of this article (10.1186/s12974-019-1558-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiangyuan Gao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Jing Z Cui
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Aikun Wang
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Hao Hang Rachel Chen
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Alison Fong
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
16
|
McCarthy CG, Wenceslau CF, Webb RC, Joe B. Novel Contributors and Mechanisms of Cellular Senescence in Hypertension-Associated Premature Vascular Aging. Am J Hypertens 2019; 32:709-719. [PMID: 30982879 DOI: 10.1093/ajh/hpz052] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 01/08/2023] Open
Abstract
Hypertension has been described as a condition of premature vascular aging, relative to actual chronological age. In fact, many factors that contribute to the deterioration of vascular function as we age are accelerated in hypertension. Nonetheless, the precise mechanisms that underlie the aged phenotype of arteries from hypertensive patients and animals remain elusive. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest. Although controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence can contribute to disease pathogenesis by presenting the senescence-associated secretory phenotype, in which molecules such as proinflammatory cytokines, matrix metalloproteases, and reactive oxygen species are released into tissue microenvironments. This review will address and critically evaluate the current literature on the role of cellular senescence in hypertension, with particular emphasis on cells types that mediate and modulate vascular function and structure.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
17
|
Moreno‐Ajona D, Chan C, Villar‐Martínez MD, Goadsby PJ. Targeting CGRP and 5‐HT
1F
Receptors for the Acute Therapy of Migraine: A Literature Review. Headache 2019; 59 Suppl 2:3-19. [DOI: 10.1111/head.13582] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 12/21/2022]
Affiliation(s)
- David Moreno‐Ajona
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience King’s College London London UK
- NIHR‐Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre King’s College Hospital London UK
| | - Calvin Chan
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience King’s College London London UK
- NIHR‐Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre King’s College Hospital London UK
| | - María Dolores Villar‐Martínez
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience King’s College London London UK
- NIHR‐Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre King’s College Hospital London UK
| | - Peter J. Goadsby
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience King’s College London London UK
- NIHR‐Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre King’s College Hospital London UK
| |
Collapse
|
18
|
Trendelenburg A, Scheuren A, Potter P, Müller R, Bellantuono I. Geroprotectors: A role in the treatment of frailty. Mech Ageing Dev 2019; 180:11-20. [DOI: 10.1016/j.mad.2019.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/25/2022]
|
19
|
Affiliation(s)
- A H Jan Danser
- From Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | - Antoinette MaassenVanDenBrink
- From Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Klee NS, McCarthy CG, Lewis S, McKenzie JL, Vincent JE, Webb RC. Urothelial Senescence in the Pathophysiology of Diabetic Bladder Dysfunction-A Novel Hypothesis. Front Surg 2018; 5:72. [PMID: 30564582 PMCID: PMC6288180 DOI: 10.3389/fsurg.2018.00072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
Abstract
Diabetic bladder dysfunction (DBD) is a well-recognized and common symptom affecting up to 50% of all diabetic patients. DBD has a broad range of clinical presentations ranging from overactive to underactive bladder symptoms that develops in middle-aged to elderly patients with long standing and poorly controlled diabetes. Low efficacy of current therapeutics and lifestyle interventions combined with high national healthcare costs highlight the need for more research into bladder dysfunction pathophysiology and novel treatment options. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest induced by replicative exhaustion and damaging insults. While controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence is known to result in tissue dysfunction through enhanced secretion of inflammatory factors. This review presents previous scientific findings and current hypotheses that characterize diabetic bladder dysfunction. Further, we propose the novel hypothesis that cellular senescence within the urothelial layer of the bladder contributes to the pro-inflammatory/pro-oxidant environment and symptoms of diabetic bladder dysfunction. Our results show increased cellular senescence in the urothelial layer of the bladder; however, whether this phenomenon is the cause or effect of DBD is unknown. The urothelial layer of the bladder is made up of transitional epithelia specialized to contract and expand with demand and plays an active role in transmission by modulating afferent activity. Transition from normal functioning urothelial cells to secretory senescence cells would not only disrupt the barrier function of this layer but may result in altered signaling and sensation of bladder fullness; dysfunction of this layer is known to result in symptoms of frequency and urgency. Future DBD therapeutics may benefit from targeting and preventing early transition of urothelial cells to senescent cells.
Collapse
Affiliation(s)
- Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Steven Lewis
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jaine L McKenzie
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States.,Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Julie E Vincent
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - R Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
21
|
Rangarajan S, Bone NB, Zmijewska AA, Jiang S, Park DW, Bernard K, Locy ML, Ravi S, Deshane J, Mannon RB, Abraham E, Darley-Usmar V, Thannickal VJ, Zmijewski JW. Metformin reverses established lung fibrosis in a bleomycin model. Nat Med 2018; 24:1121-1127. [PMID: 29967351 PMCID: PMC6081262 DOI: 10.1038/s41591-018-0087-6] [Citation(s) in RCA: 400] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 05/14/2018] [Indexed: 12/18/2022]
Abstract
Fibrosis is a pathological result of a dysfunctional repair response to tissue injury and occurs in a number of organs, including the lungs1. Cellular metabolism regulates tissue repair and remodelling responses to injury2-4. AMPK is a critical sensor of cellular bioenergetics and controls the switch from anabolic to catabolic metabolism5. However, the role of AMPK in fibrosis is not well understood. Here, we demonstrate that in humans with idiopathic pulmonary fibrosis (IPF) and in an experimental mouse model of lung fibrosis, AMPK activity is lower in fibrotic regions associated with metabolically active and apoptosis-resistant myofibroblasts. Pharmacological activation of AMPK in myofibroblasts from lungs of humans with IPF display lower fibrotic activity, along with enhanced mitochondrial biogenesis and normalization of sensitivity to apoptosis. In a bleomycin model of lung fibrosis in mice, metformin therapeutically accelerates the resolution of well-established fibrosis in an AMPK-dependent manner. These studies implicate deficient AMPK activation in non-resolving, pathologic fibrotic processes, and support a role for metformin (or other AMPK activators) to reverse established fibrosis by facilitating deactivation and apoptosis of myofibroblasts.
Collapse
Affiliation(s)
- Sunad Rangarajan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nathaniel B Bone
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna A Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shaoning Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dae Won Park
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Infectious Diseases, Korea University Ansan Hospital, Ansan, South Korea
| | - Karen Bernard
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Morgan L Locy
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saranya Ravi
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessy Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Roslyn B Mannon
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Edward Abraham
- Office of the Dean, School of Medicine, University of Miami, Miami, FL, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor J Thannickal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Jaroslaw W Zmijewski
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
22
|
Vagnoni A, Bullock SL. A cAMP/PKA/Kinesin-1 Axis Promotes the Axonal Transport of Mitochondria in Aging Drosophila Neurons. Curr Biol 2018; 28:1265-1272.e4. [PMID: 29606421 PMCID: PMC5912900 DOI: 10.1016/j.cub.2018.02.048] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 11/15/2022]
Abstract
Mitochondria play fundamental roles within cells, including energy provision, calcium homeostasis, and the regulation of apoptosis. The transport of mitochondria by microtubule-based motors is critical for neuronal structure and function. This process allows local requirements for mitochondrial functions to be met and also facilitates recycling of these organelles [1, 2]. An age-related reduction in mitochondrial transport has been observed in neurons of mammalian and non-mammalian organisms [3, 4, 5, 6], and has been proposed to contribute to the broader decline in neuronal function that occurs during aging [3, 5, 6, 7]. However, the factors that influence mitochondrial transport in aging neurons are poorly understood. Here we provide evidence using the tractable Drosophila wing nerve system that the cyclic AMP/protein kinase A (cAMP/PKA) pathway promotes the axonal transport of mitochondria in adult neurons. The level of the catalytic subunit of PKA decreases during aging, and acute activation of the cAMP/PKA pathway in aged flies strongly stimulates mitochondrial motility. Thus, the age-related impairment of transport is reversible. The expression of many genes is increased by PKA activation in aged flies. However, our results indicate that elevated mitochondrial transport is due in part to upregulation of the heavy chain of the kinesin-1 motor, the level of which declines during aging. Our study identifies evolutionarily conserved factors that can strongly influence mitochondrial motility in aging neurons. cAMP/PKA pathway promotes mitochondrial transport in adult Drosophila wing neurons Pathway activation in aged flies suppresses age-related reduction in transport Levels of PKAc and kinesin-1 motor decline during aging Kinesin-1 upregulation is an important output of PKA activation in aged flies
Collapse
Affiliation(s)
- Alessio Vagnoni
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London SE5 9RX, UK.
| | - Simon L Bullock
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
23
|
Abstract
Numerous approaches have been taken in the hunt for human disease genes. The identification of such genes not only provides a great deal of information about the mechanism of disease development, but also provides potential avenues for better diagnosis and treatment. In this chapter, we review the use of the nonmammalian model organism C. elegans for the identification of human disease genes. Studies utilizing this relatively simple organism offer a good balance between the ability to recapitulate many aspects of human disease, while still offering an abundance of powerful cell biological, genetic, and genomic tools for disease gene discovery. C. elegans and other nonmammalian models have produced, and will continue to produce, key insights into human disease pathogenesis.
Collapse
Affiliation(s)
- Javier Apfeld
- Department of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Scott Alper
- Department of Biomedical Research, Center for Genes, Environment and Health, National Jewish Health, Denver, CO, 80206, USA.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
24
|
Bajwa P, Nagendra PB, Nielsen S, Sahoo SS, Bielanowicz A, Lombard JM, Wilkinson JE, Miller RA, Tanwar PS. Age related increase in mTOR activity contributes to the pathological changes in ovarian surface epithelium. Oncotarget 2017; 7:19214-27. [PMID: 27036037 PMCID: PMC4991377 DOI: 10.18632/oncotarget.8468] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/23/2016] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is a disease of older women. However, the molecular mechanisms of ovarian aging and their contribution to the pathogenesis of ovarian cancer are currently unclear. mTOR signalling is a major regulator of aging as suppression of this pathway extends lifespan in model organisms. Overactive mTOR signalling is present in up to 80% of ovarian cancer samples and is associated with poor prognosis. This study examined the role of mTOR signalling in age-associated changes in ovarian surface epithelium (OSE). Histological examination of ovaries from both aged mice and women revealed OSE cell hyperplasia, papillary growth and inclusion cysts. These pathological lesions expressed bonafide markers of ovarian cancer precursor lesions, Pax8 and Stathmin 1, and were presented with elevated mTOR signalling. To understand whether overactive mTOR signalling is responsible for the development of these pathological changes, we analysed ovaries of the Pten trangenic mice and found significant reduction in OSE lesions compared to controls. Furthermore, pharmacological suppression of mTOR signalling significantly decreased OSE hyperplasia in aged mice. Treatment with mTOR inhibitors reduced human ovarian cancer cell viability, proliferation and colony forming ability. Collectively, we have established the role of mTOR signalling in age-related OSE pathologies and initiation of ovarian cancer.
Collapse
Affiliation(s)
- Preety Bajwa
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, New South Wales, Australia
| | - Prathima B Nagendra
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, New South Wales, Australia
| | | | - Subhransu S Sahoo
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, New South Wales, Australia
| | - Amanda Bielanowicz
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, New South Wales, Australia
| | - Janine M Lombard
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia.,Division of Gynaecology Oncology, Department of Medical Oncology, Calvary Mater Newcastle, Waratah, New South Wales, Australia
| | - J Erby Wilkinson
- Unit for Laboratory Animal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Pradeep S Tanwar
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, New South Wales, Australia
| |
Collapse
|
25
|
Ferrosenescence: The iron age of neurodegeneration? Mech Ageing Dev 2017; 174:63-75. [PMID: 29180225 DOI: 10.1016/j.mad.2017.11.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Aging has been associated with iron retention in many cell types, including the neurons, promoting neurodegeneration by ferroptosis. Excess intracellular iron accelerates aging by damaging the DNA and blocking genomic repair systems, a process we define as ferrosenescence. Novel neuroimaging and proteomic techniques have pinpointed indicators of both iron retention and ferrosenescence, allowing for their early correction, potentially bringing prevention of neurodegenerative disorders within reach. In this review, we take a closer look at the early markers of iron dyshomeostasis in neurodegenerative disorders, focusing on preventive strategies based on nutritional and microbiome manipulations.
Collapse
|
26
|
Gao AW, Uit de Bos J, Sterken MG, Kammenga JE, Smith RL, Houtkooper RH. Forward and reverse genetics approaches to uncover metabolic aging pathways in Caenorhabditis elegans. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2697-2706. [PMID: 28919364 DOI: 10.1016/j.bbadis.2017.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 01/08/2023]
Abstract
The biological mechanisms of aging have been studied in depth and prominent findings in this field promote the development of new therapies for age-associated disorders. Various model organisms are used for research on aging; among these, the nematode Caenorhabditis elegans has been widely used and has provided valuable knowledge in determining the regulatory mechanisms driving the aging process. Many genes involved in lifespan regulation are associated with metabolic pathways and are influenced by genetic and environmental factors. In line with this, C. elegans provides a promising platform to study such gene by environment interactions, in either a reverse or forward genetics approach. In this review, we discuss longevity mechanisms related to metabolic networks that have been discovered in C. elegans. We also highlight the use of wild populations to study the complex genetic basis of natural variation for quantitative traits that mediate longevity.
Collapse
Affiliation(s)
- Arwen W Gao
- Laboratory Genetic Metabolic Diseases, Academic Medical Center of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jelmi Uit de Bos
- Laboratory Genetic Metabolic Diseases, Academic Medical Center of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Mark G Sterken
- Laboratory of Nematology, Wageningen University and Research, 6708 PB Wageningen, The Netherlands
| | - Jan E Kammenga
- Laboratory of Nematology, Wageningen University and Research, 6708 PB Wageningen, The Netherlands
| | - Reuben L Smith
- Laboratory Genetic Metabolic Diseases, Academic Medical Center of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Academic Medical Center of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
27
|
François JC, Aïd S, Chaker Z, Lacube P, Xu J, Fayad R, Côté F, Even P, Holzenberger M. Disrupting IGF Signaling in Adult Mice Conditions Leanness, Resilient Energy Metabolism, and High Growth Hormone Pulses. Endocrinology 2017; 158:2269-2283. [PMID: 28881863 DOI: 10.1210/en.2017-00261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/05/2017] [Indexed: 12/26/2022]
Abstract
Growth hormone (GH) and insulinlike growth factor (IGF) promote aging and age-related pathologies. Inhibiting this pathway by targeting IGF receptor (IGF-1R) is a promising strategy to extend life span, alleviate age-related diseases, and reduce tumor growth. Although anti-IGF-1R agents are being developed, long-term effects of IGF-1R blockade remain unknown. In this study, we used ubiquitous inducible IGF-1R knockout (UBIKOR) to suppress signaling in all adult tissues and screened health extensively. Surprisingly, UBIKOR mice showed no overt defects and presented with rather inconspicuous health, including normal cognition. Endocrine GH and IGF-1 were strongly upregulated without causing acromegaly. UBIKOR mice were strikingly lean with coordinate changes in body composition and organ size. They were insulin resistant but preserved physiological energy expenditure and displayed enhanced fasting metabolic flexibility. Thus, long-term IGF-1R blockade generated beneficial effects on aging-relevant metabolism, but exposed to high GH. This needs to be considered when targeting IGF-1R to protect from neurodegeneration, retard aging, or fight cancer.
Collapse
Affiliation(s)
| | - Saba Aïd
- INSERM Research Center Unité 938, 75012 Paris, France
- Sorbonne University, 75005 Paris, France
| | - Zayna Chaker
- INSERM Research Center Unité 938, 75012 Paris, France
- Faculty of Medicine, University Paris Descartes, 75006 Paris, France
| | | | - Jie Xu
- INSERM Research Center Unité 938, 75012 Paris, France
- Sorbonne University, 75005 Paris, France
| | - Racha Fayad
- INSERM Research Center Unité 938, 75012 Paris, France
- Faculty of Medicine, University Paris Descartes, 75006 Paris, France
| | - Francine Côté
- Institut Imagine INSERM Unité 1163/CNRS Equipe 8254, Necker Enfants Malades Hospital, 75015 Paris, France
| | - Patrick Even
- AgroParisTech, INRA, Université Paris Saclay, Nutrition Physiology and Ingestive Behavior Unité 914, 75005 Paris, France
| | - Martin Holzenberger
- INSERM Research Center Unité 938, 75012 Paris, France
- Sorbonne University, 75005 Paris, France
| |
Collapse
|
28
|
Corrêa RCG, Peralta RM, Haminiuk CWI, Maciel GM, Bracht A, Ferreira ICFR. New phytochemicals as potential human anti-aging compounds: Reality, promise, and challenges. Crit Rev Food Sci Nutr 2017; 58:942-957. [DOI: 10.1080/10408398.2016.1233860] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rúbia C. G. Corrêa
- Graduate Program in Food Science, State University of Maringá (UEM), Paraná, Brazil
- Graduate Program in Food Technology, Federal Technological University of Paraná (UTFPR), Campus Campo Mourão, Paraná, Brazil
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| | - Rosane M. Peralta
- Graduate Program in Food Science, State University of Maringá (UEM), Paraná, Brazil
- Department of Biochemistry, State University of Maringá (UEM), Paraná, Brazil
- Mountain Research Centre (CIMO), ESA, Polytechnic Institute of Bragança (IPB), Campus de Santa Apolónia, Bragança, Portugal
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| | - Charles W. I. Haminiuk
- Graduate Program in Food Technology, Federal Technological University of Paraná (UTFPR), Campus Campo Mourão, Paraná, Brazil
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| | - Giselle Maria Maciel
- Mountain Research Centre (CIMO), ESA, Polytechnic Institute of Bragança (IPB), Campus de Santa Apolónia, Bragança, Portugal
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| | - Adelar Bracht
- Graduate Program in Food Science, State University of Maringá (UEM), Paraná, Brazil
- Department of Biochemistry, State University of Maringá (UEM), Paraná, Brazil
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| | - Isabel C. F. R. Ferreira
- Mountain Research Centre (CIMO), ESA, Polytechnic Institute of Bragança (IPB), Campus de Santa Apolónia, Bragança, Portugal
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná (UTFPR), Curitiba, Paraná, Brazil
| |
Collapse
|
29
|
Shiels PG, McGuinness D, Eriksson M, Kooman JP, Stenvinkel P. The role of epigenetics in renal ageing. Nat Rev Nephrol 2017. [PMID: 28626222 DOI: 10.1038/nrneph.2017.78] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An ability to separate natural ageing processes from processes specific to morbidities is required to understand the heterogeneity of age-related organ dysfunction. Mechanistic insight into how epigenetic factors regulate ageing throughout the life course, linked to a decline in renal function with ageing, is already proving to be of value in the analyses of clinical and epidemiological cohorts. Noncoding RNAs provide epigenetic regulatory circuits within the kidney, which reciprocally interact with DNA methylation processes, histone modification and chromatin. These interactions have been demonstrated to reflect the biological age and function of renal allografts. Epigenetic factors control gene expression and activity in response to environmental perturbations. They also have roles in highly conserved signalling pathways that modulate ageing, including the mTOR and insulin/insulin-like growth factor signalling pathways, and regulation of sirtuin activity. Nutrition, the gut microbiota, inflammation and environmental factors, including psychosocial and lifestyle stresses, provide potential mechanistic links between the epigenetic landscape of ageing and renal dysfunction. Approaches to modify the renal epigenome via nutritional intervention, targeting the methylome or targeting chromatin seem eminently feasible, although caution is merited owing to the potential for intergenerational and transgenerational effects.
Collapse
Affiliation(s)
- Paul G Shiels
- Section of Epigenetics, Institute of Cancer Sciences, Wolfson Wohl Translational Research Centre, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Dagmara McGuinness
- Section of Epigenetics, Institute of Cancer Sciences, Wolfson Wohl Translational Research Centre, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Maria Eriksson
- Department of Biosciences and Nutrition (BioNut), H2, Eriksson, Novum 141, 83 Huddinge, Sweden
| | - Jeroen P Kooman
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastrich, Netherlands
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Huddinge, Karolinska Institutet, SE-14157 Stockholm, Sweden
| |
Collapse
|
30
|
Abstract
The health of an organism is orchestrated by a multitude of molecular and biochemical networks responsible for ensuring homeostasis within cells and tissues. However, upon aging, a progressive failure in the maintenance of this homeostatic balance occurs in response to a variety of endogenous and environmental stresses, allowing the accumulation of damage, the physiological decline of individual tissues, and susceptibility to diseases. What are the molecular and cellular signaling events that control the aging process and how can this knowledge help design therapeutic strategies to combat age-associated diseases? Here we provide a comprehensive overview of the evolutionarily conserved biological processes that alter the rate of aging and discuss their link to disease prevention and the extension of healthy life span.
Collapse
Affiliation(s)
- Celine E Riera
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720; .,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815.,Glenn Center for Research on Aging, University of California, Berkeley, California 94720
| | - Carsten Merkwirth
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720; .,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815.,Glenn Center for Research on Aging, University of California, Berkeley, California 94720
| | - C Daniel De Magalhaes Filho
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815.,The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Andrew Dillin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720; .,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815.,Glenn Center for Research on Aging, University of California, Berkeley, California 94720
| |
Collapse
|
31
|
Cui H, Ge J, Xie N, Banerjee S, Zhou Y, Liu RM, Thannickal VJ, Liu G. miR-34a promotes fibrosis in aged lungs by inducing alveolarepithelial dysfunctions. Am J Physiol Lung Cell Mol Physiol 2016; 312:L415-L424. [PMID: 27979858 DOI: 10.1152/ajplung.00335.2016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/28/2016] [Accepted: 12/12/2016] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a well-known age-related disease. However, much less recognized has been the aging associated pathogenesis of this disorder. As we and others previously showed that dysregulation of micro-RNAs (miRNAs) was an important mechanism involved in pulmonary fibrosis, the role of these molecules in this pathology in the aged population has not been investigated (Cushing L, Kuang PP, Qian J, Shao F, Wu J, Little F, Thannickal VJ, Cardoso WV, Lü J. Am J Respir Cell Mol Biol 45: 287-294, 2011; Liu G, Friggeri A, Yang Y, Milosevic J, Ding Q, Thannickal VJ, Kaminski N, Abraham E. J Exp Med 207: 1589-1597, 2010; Pandit KV, Corcoran D, Yousef H, Yarlagadda M, Tzouvelekis A, Gibson KF, Konishi K, Yousem SA, Singh M, Handley D, Richards T, Selman M, Watkins SC, Pardo A, Ben-Yehudah A, Bouros D, Eickelberg O, Ray P, Benos PV, Kaminski N. Am J Respir Crit Care Med 182: 220-229, 2010). In this study, by using a lung fibrosis model established in old mice, we found that ablation of miR-34a protected aged animals from developing experimental lung fibrosis. miR-34a was upregulated in lung epithelial cells, but not in lung fibroblasts of aged mice, and miR-34a expression was further increased in epithelial cells of the fibrotic lungs of these old animals. We found that miR-34a induced dysfunctions in alveolar epithelial cells (AECs), as evidenced by increased cellular senescence and apoptosis and mitochondrial aberrations. More importantly, these abnormalities were attenuated in AECs of the fibrotic lungs of aged miR-34a-/- mice. We found that miR-34a targeted Sirt1, a master anti-aging regulator, and two key cell cycle modulators, E2F3 and cyclin E2, in lung epithelial cells, and the repression of these targets was relieved in miR-34a-deficient AECs. In summary, our data suggest that elevated AEC miR-34a plays a critical role in the pathogenesis of pulmonary fibrosis in the aged population. Our study also indicates miR-34a to be a more precise miRNA target for treating this disease that overwhelmingly affects people of advanced age.
Collapse
Affiliation(s)
- Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Jing Ge
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and.,Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Na Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Yong Zhou
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
32
|
Figueira I, Fernandes A, Mladenovic Djordjevic A, Lopez-Contreras A, Henriques CM, Selman C, Ferreiro E, Gonos ES, Trejo JL, Misra J, Rasmussen LJ, Xapelli S, Ellam T, Bellantuono I. Interventions for age-related diseases: Shifting the paradigm. Mech Ageing Dev 2016; 160:69-92. [DOI: 10.1016/j.mad.2016.09.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/18/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
|
33
|
MaassenVanDenBrink A, Meijer J, Villalón CM, Ferrari MD. Wiping Out CGRP: Potential Cardiovascular Risks. Trends Pharmacol Sci 2016; 37:779-788. [DOI: 10.1016/j.tips.2016.06.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 01/06/2023]
|
34
|
|
35
|
Castillo-Quan JI, Li L, Kinghorn KJ, Ivanov DK, Tain LS, Slack C, Kerr F, Nespital T, Thornton J, Hardy J, Bjedov I, Partridge L. Lithium Promotes Longevity through GSK3/NRF2-Dependent Hormesis. Cell Rep 2016; 15:638-650. [PMID: 27068460 PMCID: PMC4850359 DOI: 10.1016/j.celrep.2016.03.041] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/31/2016] [Accepted: 03/10/2016] [Indexed: 01/06/2023] Open
Abstract
The quest to extend healthspan via pharmacological means is becoming increasingly urgent, both from a health and economic perspective. Here we show that lithium, a drug approved for human use, promotes longevity and healthspan. We demonstrate that lithium extends lifespan in female and male Drosophila, when administered throughout adulthood or only later in life. The life-extending mechanism involves the inhibition of glycogen synthase kinase-3 (GSK-3) and activation of the transcription factor nuclear factor erythroid 2-related factor (NRF-2). Combining genetic loss of the NRF-2 repressor Kelch-like ECH-associated protein 1 (Keap1) with lithium treatment revealed that high levels of NRF-2 activation conferred stress resistance, while low levels additionally promoted longevity. The discovery of GSK-3 as a therapeutic target for aging will likely lead to more effective treatments that can modulate mammalian aging and further improve health in later life.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Li Li
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Kerri J Kinghorn
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Dobril K Ivanov
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Luke S Tain
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Cathy Slack
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Fiona Kerr
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Tobias Nespital
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Janet Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Ivana Bjedov
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Linda Partridge
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany.
| |
Collapse
|
36
|
Barton M, Husmann M, Meyer MR. Accelerated Vascular Aging as a Paradigm for Hypertensive Vascular Disease: Prevention and Therapy. Can J Cardiol 2016; 32:680-686.e4. [PMID: 27118295 DOI: 10.1016/j.cjca.2016.02.062] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 12/21/2022] Open
Abstract
Aging is considered the most important nonmodifiable risk factor for cardiovascular disease and death after age 28 years. Because of demographic changes the world population is expected to increase to 9 billion by the year 2050 and up to 12 billion by 2100, with several-fold increases among those 65 years of age and older. Healthy aging and prevention of aging-related diseases and associated health costs have become part of political agendas of governments around the world. Atherosclerotic vascular burden increases with age; accordingly, patients with progeria (premature aging) syndromes die from myocardial infarctions or stroke as teenagers or young adults. The incidence and prevalence of arterial hypertension also increases with age. Arterial hypertension-like diabetes and chronic renal failure-shares numerous pathologies and underlying mechanisms with the vascular aging process. In this article, we review how arterial hypertension resembles premature vascular aging, including the mechanisms by which arterial hypertension (as well as other risk factors such as diabetes mellitus, dyslipidemia, or chronic renal failure) accelerates the vascular aging process. We will also address the importance of cardiovascular risk factor control-including antihypertensive therapy-as a powerful intervention to interfere with premature vascular aging to reduce the age-associated prevalence of diseases such as myocardial infarction, heart failure, hypertensive nephropathy, and vascular dementia due to cerebrovascular disease. Finally, we will discuss the implementation of endothelial therapy, which aims at active patient participation to improve primary and secondary prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
| | - Marc Husmann
- Division of Angiology, University Hospital Zürich, Zürich, Switzerland
| | | |
Collapse
|
37
|
Irwin MH, Moos WH, Faller DV, Steliou K, Pinkert CA. Epigenetic Treatment of Neurodegenerative Disorders: Alzheimer and Parkinson Diseases. Drug Dev Res 2016; 77:109-23. [PMID: 26899010 DOI: 10.1002/ddr.21294] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preclinical Research In this review, we discuss epigenetic-driven methods for treating neurodegenerative disorders associated with mitochondrial dysfunction, focusing on carnitinoid antioxidant-histone deacetylase inhibitors that show an ability to reinvigorate synaptic plasticity and protect against neuromotor decline in vivo. Aging remains a major risk factor in patients who progress to dementia, a clinical syndrome typified by decreased mental capacity, including impairments in memory, language skills, and executive function. Energy metabolism and mitochondrial dysfunction are viewed as determinants in the aging process that may afford therapeutic targets for a host of disease conditions, the brain being primary in such thinking. Mitochondrial dysfunction is a core feature in the pathophysiology of both Alzheimer and Parkinson diseases and rare mitochondrial diseases. The potential of new therapies in this area extends to glaucoma and other ophthalmic disorders, migraine, Creutzfeldt-Jakob disease, post-traumatic stress disorder, systemic exertion intolerance disease, and chemotherapy-induced cognitive impairment. An emerging and hopefully more promising approach to addressing these hard-to-treat diseases leverages their sensitivity to activation of master regulators of antioxidant and cytoprotective genes, antioxidant response elements, and mitophagy. Drug Dev Res 77 : 109-123, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.,SRI Biosciences, A Division of SRI International, Menlo Park, CA, USA
| | - Douglas V Faller
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA.,PhenoMatriX, Inc., Boston, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.,Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|