1
|
Lau V, Awogbindin I, Tremblay MÈ. Chronic stress induces senescence build-up early in life. NATURE AGING 2025:10.1038/s43587-024-00774-1. [PMID: 39747653 DOI: 10.1038/s43587-024-00774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Victor Lau
- University of Victoria, Victoria, British Columbia, Canada
| | | | | |
Collapse
|
2
|
Mansfield L, Ramponi V, Gupta K, Stevenson T, Mathew AB, Barinda AJ, Herbstein F, Morsli S. Emerging insights in senescence: pathways from preclinical models to therapeutic innovations. NPJ AGING 2024; 10:53. [PMID: 39578455 PMCID: PMC11584693 DOI: 10.1038/s41514-024-00181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Senescence is a crucial hallmark of ageing and a significant contributor to the pathology of age-related disorders. As committee members of the young International Cell Senescence Association (yICSA), we aim to synthesise recent advancements in the identification, characterisation, and therapeutic targeting of senescence for clinical translation. We explore novel molecular techniques that have enhanced our understanding of senescent cell heterogeneity and their roles in tissue regeneration and pathology. Additionally, we delve into in vivo models of senescence, both non-mammalian and mammalian, to highlight tools available for advancing the contextual understanding of in vivo senescence. Furthermore, we discuss innovative diagnostic tools and senotherapeutic approaches, emphasising their potential for clinical application. Future directions of senescence research are explored, underscoring the need for precise, context-specific senescence classification and the integration of advanced technologies such as machine learning, long-read sequencing, and multifunctional senoprobes and senolytics. The dual role of senescence in promoting tissue homoeostasis and contributing to chronic diseases highlights the complexity of targeting these cells for improved clinical outcomes.
Collapse
Affiliation(s)
- Luke Mansfield
- The Bateson Centre, School of Medicine and Population Health, The University of Sheffield, Western Bank, Sheffield, UK
| | - Valentina Ramponi
- Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Kavya Gupta
- Department of Cellular and Molecular Biology and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Abraham Binoy Mathew
- Department of Developmental Biology and Genetics, Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Agian Jeffilano Barinda
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Metabolic, Cardiovascular, and Aging Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Florencia Herbstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - Samir Morsli
- Karolinska Institutet, Department of Cell and Molecular Biology, Biomedicum Q6A, Stockholm, Sweden.
| |
Collapse
|
3
|
Ávila BM, Zanini BM, Luduvico KP, Oliveira TL, Hense JD, Garcia DN, Prosczek J, Stefanello FM, da Cruz PH, Giongo JL, Vaucher RA, Mason JB, Masternak MM, Schneider A. Effect of senolytic drugs in young female mice chemically induced to estropause. Life Sci 2024; 357:123073. [PMID: 39307182 DOI: 10.1016/j.lfs.2024.123073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
AIMS This study aimed to assess metabolic responses and senescent cell burden in young female mice induced to estropause and treated with senolytic drugs. MAIN METHODS Estropause was induced by 4-vinylcyclohexene diepoxide (VCD) injection in two-month-old mice. The senolytics dasatinib and quercetin (D + Q) or fisetin were given by oral gavage once a month from five to 11 months of age. KEY FINDINGS VCD-induced estropause led to increased body mass and reduced albumin concentrations compared to untreated cyclic mice, without affecting insulin sensitivity, lipid profile, liver enzymes, or total proteins. Estropause decreased catalase activity in adipose tissue but had no significant effect on other redox parameters in adipose and hepatic tissues. Fisetin treatment reduced ROS levels in the hepatic tissue of estropause mice. Estropause did not influence senescence-associated beta-galactosidase activity in adipose and hepatic tissues but increased senescent cell markers and fibrosis in ovaries. Senolytic treatment did not decrease ovarian cellular senescence induced by estropause. SIGNIFICANCE Overall, the findings suggest that estropause leads to minor metabolic changes in young females, and the senolytics D + Q and fisetin had no protective effects despite increased ovarian senescence.
Collapse
Affiliation(s)
- Bianca M Ávila
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Bianka M Zanini
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Karina P Luduvico
- Center of Chemical, Pharmaceutical, and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Thais L Oliveira
- Biotechnology Center, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jéssica D Hense
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Juliane Prosczek
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Francieli M Stefanello
- Center of Chemical, Pharmaceutical, and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Pedro H da Cruz
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Janice L Giongo
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rodrigo A Vaucher
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeffrey B Mason
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
4
|
Roberts H, Fang Y, Quinn K, Hill T, Peck MR, Bartke A, Hascup KN, Hascup ER. Lifespan of male and female APP/PS1 and APP NL-F/NL-F mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618508. [PMID: 39464050 PMCID: PMC11507819 DOI: 10.1101/2024.10.15.618508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) disproportionately affects women, yet most preclinical research studies are male-centric. We performed lifespan analyses of male and female AD mouse models (APP/PS1 and APPNL-F/NL-F) and their shared genetic background control (C57BL/6). Survival curves support significant sex differences between within genotypes. Minimal longevity revealed increased age in male APP/PS1, and decreased age in APPNL-F/NL-F mice. Maximal longevity revealed an increased average age in males. Furthermore, median lifespan differed between sex and genotype. This study supports sexual dimorphic survival in two mouse models of AD, emphasizing the need to examine mechanisms and treatments in both sexes.
Collapse
Affiliation(s)
- Hannah Roberts
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Yimin Fang
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Tiarra Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Dept of Internal Medicine; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
5
|
Tavenier J, Nehlin JO, Houlind MB, Rasmussen LJ, Tchkonia T, Kirkland JL, Andersen O, Rasmussen LJH. Fisetin as a senotherapeutic agent: Evidence and perspectives for age-related diseases. Mech Ageing Dev 2024; 222:111995. [PMID: 39384074 DOI: 10.1016/j.mad.2024.111995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Fisetin, a flavonoid naturally occurring in plants, fruits, and vegetables, has recently gained attention for its potential role as a senotherapeutic agent for the treatment of age-related chronic diseases. Senotherapeutics target senescent cells, which accumulate with age and disease, in both circulating immune cell populations and solid organs and tissues. Senescent cells contribute to development of many chronic diseases, primarily by eliciting systemic chronic inflammation through their senescence-associated secretory phenotype. Here, we explore whether fisetin as a senotherapeutic can eliminate senescent cells, and thereby alleviate chronic diseases, by examining current evidence from in vitro studies and animal models that investigate fisetin's impact on age-related diseases, as well as from phase I/II trials in various patient populations. We discuss the application of fisetin in humans, including challenges and future directions. Our review of available data suggests that targeting senescent cells with fisetin offers a promising strategy for managing multiple chronic diseases, potentially transforming future healthcare for older and multimorbid patients. However, further studies are needed to establish the safety, pharmacokinetics, and efficacy of fisetin as a senotherapeutic, identify relevant and reliable outcome measures in human trials, optimize dosing, and better understand the possible limitations of fisetin as a senotherapeutic agent.
Collapse
Affiliation(s)
- Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; The Hospital Pharmacy, Marielundsvej 25, Herlev 2730, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| | - Tamara Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N 2200, Denmark; The Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Psychology & Neuroscience, Duke University, 2020 West Main Street Suite 201, Durham, NC 27708, USA.
| |
Collapse
|
6
|
Maher P. The flavonoid fisetin reduces multiple physiological risk factors for dementia. Neurochem Int 2024; 178:105805. [PMID: 39004102 DOI: 10.1016/j.neuint.2024.105805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Dementia is a growing problem around the globe as the world's population continues to age. Multiple studies have identified potentially modifiable risk factors for the development of dementia suggesting that addressing some or all of these risk factors might have a significant impact on the aging population worldwide. However, this is not always as straightforward as it seems since many of these risk factors are currently treated with drugs specific to the risk factor. Moreover, since people can have multiple risk factors, addressing each of them individually could be highly problematic as it would likely lead to negative outcomes associated with polypharmacy and, in the long term, could do significant harm. A potential alternative is to identify compounds that have shown efficacy against a number of these different risk factors. As discussed in this review, there is strong evidence that the flavonol fisetin is one such compound. In animal studies it has shown efficacy against many of the risk factors that have been associated with an increased risk of developing dementia and also exhibits direct neuroprotective effects. Thus, further human research on fisetin in the context of dementia risk factors is clearly warranted.
Collapse
Affiliation(s)
- Pamela Maher
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
7
|
Fang Y, Peck MR, Quinn K, Chapman JE, Medina D, McFadden SA, Bartke A, Hascup ER, Hascup KN. Senolytic intervention improves cognition, metabolism, and adiposity in female APP NL-F/NL-F mice. GeroScience 2024:10.1007/s11357-024-01308-8. [PMID: 39120687 DOI: 10.1007/s11357-024-01308-8] [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: 03/15/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Senescent cells accumulate throughout the body and brain contributing to unhealthy aging and Alzheimer's disease (AD). The APPNL-F/NL-F amyloidogenic AD mouse model exhibits increased markers of senescent cells and the senescence-associated secretory phenotype (SASP) in visceral white adipose tissue and the hippocampus before plaque accumulation and cognitive decline. We hypothesized that senolytic intervention would alleviate cellular senescence thereby improving spatial memory in APPNL-F/NL-F mice. Thus, 4-month-old male and female APPNL-F/NL-F mice were treated monthly with vehicle, 5 mg/kg dasatinib + 50 mg/kg quercetin, or 100 mg/kg fisetin. Blood glucose levels, energy metabolism, spatial memory, amyloid burden, and senescent cell markers were assayed. Dasatinib + quercetin treatment in female APPNL-F/NL-F mice increased oxygen consumption and energy expenditure resulting in decreased body mass. White adipose tissue mass was decreased along with senescence markers, SASP, blood glucose, and plasma insulin and triglycerides. Hippocampal senescence markers and SASP were reduced along with soluble and insoluble amyloid-β (Aβ)42 and senescence-associated-β-gal activity leading to improved spatial memory. Fisetin had negligible effects on these measures in female APPNL-F/NL-F mice while neither senolytic intervention altered these parameters in the male mice. Considering women have a greater risk of dementia, identifying senotherapeutics appropriate for sex and disease stage is necessary for personalized medicine.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Mackenzie R Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Jenelle E Chapman
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Samuel A McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
8
|
Hense JD, Garcia DN, Zanini BM, Barreto MM, Perreira GC, Isola JVV, de Brito C, Fornalik M, Mondal SA, Ávila BM, Oliveira TL, Rice HC, Lacy CI, Vaucher RA, Mason JB, Masternak MM, Stout MB, Schneider A. MASLD does not affect fertility and senolytics fail to prevent MASLD progression in male mice. Sci Rep 2024; 14:17332. [PMID: 39068167 PMCID: PMC11283523 DOI: 10.1038/s41598-024-67697-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
Senescent cells have been linked to the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). However, the effectiveness of senolytic drugs in reducing liver damage in mice with MASLD is not clear. Additionally, MASLD has been reported to adversely affect male reproductive function. Therefore, this study aimed to evaluate the protective effect of senolytic drugs on liver damage and fertility in male mice with MASLD. Three-month-old male mice were fed a standard diet (SD) or a choline-deficient western diet (WD) until 9 months of age. At 6 months of age mice were randomized within dietary treatment groups into senolytic (dasatinib + quercetin [D + Q]; fisetin [FIS]) or vehicle control treatment groups. We found that mice fed choline-deficient WD had liver damage characteristic of MASLD, with increased liver size, triglycerides accumulation, fibrosis, along increased liver cellular senescence and liver and systemic inflammation. Senolytics were not able to reduce liver damage, senescence and systemic inflammation, suggesting limited efficacy in controlling WD-induced liver damage. Sperm quality and fertility remained unchanged in mice developing MASLD or receiving senolytics. Our data suggest that liver damage and senescence in mice developing MASLD is not reversible by the use of senolytics. Additionally, neither MASLD nor senolytics affected fertility in male mice.
Collapse
Affiliation(s)
- Jessica D Hense
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Bianka M Zanini
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Mariana M Barreto
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Giulia C Perreira
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Camila de Brito
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Michal Fornalik
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Samim A Mondal
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
- Department of Endocrinology, JIPMER, Puducherry, 605006, India
| | - Bianca M Ávila
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Thais L Oliveira
- Biotechnology Center, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Heather C Rice
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Charles I Lacy
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Rodrigo A Vaucher
- Center for Chemical, Pharmaceutical and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeffrey B Mason
- Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil.
| |
Collapse
|
9
|
Hense JD, Isola JVV, Garcia DN, Magalhães LS, Masternak MM, Stout MB, Schneider A. The role of cellular senescence in ovarian aging. NPJ AGING 2024; 10:35. [PMID: 39033161 PMCID: PMC11271274 DOI: 10.1038/s41514-024-00157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 07/23/2024]
Abstract
This review explores the relationship between ovarian aging and senescent cell accumulation, as well as the efficacy of senolytics to improve reproductive longevity. Reproductive longevity is determined by the age-associated decline in ovarian reserve, resulting in reduced fertility and eventually menopause. Cellular senescence is a state of permanent cell cycle arrest and resistance to apoptosis. Senescent cells accumulate in several tissues with advancing age, thereby promoting chronic inflammation and age-related diseases. Ovaries also appear to accumulate senescent cells with age, which might contribute to aging of the reproductive system and whole organism through SASP production. Importantly, senolytic drugs can eliminate senescent cells and may present a potential intervention to mitigate ovarian aging. Herein, we review the current literature related to the efficacy of senolytic drugs for extending the reproductive window in mice.
Collapse
Affiliation(s)
- Jéssica D Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | | | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
10
|
Liang Y, Kaushal D, Wilson RB. Cellular Senescence and Extracellular Vesicles in the Pathogenesis and Treatment of Obesity-A Narrative Review. Int J Mol Sci 2024; 25:7943. [PMID: 39063184 PMCID: PMC11276987 DOI: 10.3390/ijms25147943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This narrative review explores the pathophysiology of obesity, cellular senescence, and exosome release. When exposed to excessive nutrients, adipocytes develop mitochondrial dysfunction and generate reactive oxygen species with DNA damage. This triggers adipocyte hypertrophy and hypoxia, inhibition of adiponectin secretion and adipogenesis, increased endoplasmic reticulum stress and maladaptive unfolded protein response, metaflammation, and polarization of macrophages. Such feed-forward cycles are not resolved by antioxidant systems, heat shock response pathways, or DNA repair mechanisms, resulting in transmissible cellular senescence via autocrine, paracrine, and endocrine signaling. Senescence can thus affect preadipocytes, mature adipocytes, tissue macrophages and lymphocytes, hepatocytes, vascular endothelium, pancreatic β cells, myocytes, hypothalamic nuclei, and renal podocytes. The senescence-associated secretory phenotype is closely related to visceral adipose tissue expansion and metaflammation; inhibition of SIRT-1, adiponectin, and autophagy; and increased release of exosomes, exosomal micro-RNAs, pro-inflammatory adipokines, and saturated free fatty acids. The resulting hypernefemia, insulin resistance, and diminished fatty acid β-oxidation lead to lipotoxicity and progressive obesity, metabolic syndrome, and physical and cognitive functional decline. Weight cycling is related to continuing immunosenescence and exposure to palmitate. Cellular senescence, exosome release, and the transmissible senescence-associated secretory phenotype contribute to obesity and metabolic syndrome. Targeted therapies have interrelated and synergistic effects on cellular senescence, obesity, and premature aging.
Collapse
Affiliation(s)
- Yicong Liang
- Bankstown Hospital, University of New South Wales, Sydney, NSW 2560, Australia;
| | - Devesh Kaushal
- Campbelltown Hospital, Western Sydney University, Sydney, NSW 2560, Australia;
| | - Robert Beaumont Wilson
- School of Clinical Medicine, University of New South Wales, High St., Kensington, Sydney, NSW 2052, Australia
| |
Collapse
|
11
|
Fang Y, Peck MR, Quinn K, Chapman JE, Medina D, McFadden SA, Bartke A, Hascup KN, Hascup ER. Senolytic Intervention Improves Cognition, Metabolism, and Adiposity in Female APP NL-F/NL-F Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.12.571277. [PMID: 38168356 PMCID: PMC10760014 DOI: 10.1101/2023.12.12.571277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Senescent cells accumulate throughout the body and brain contributing to unhealthy aging and Alzheimer's disease (AD). The APP NL-F/NL-F amyloidogenic AD mouse model exhibits increased markers of senescent cells and the senescence-associated secretory phenotype (SASP) in visceral white adipose tissue before plaque accumulation and cognitive decline. We hypothesized that senolytic intervention would alleviate cellular senescence thereby improving spatial memory in APP NL-F/NL-F mice. Thus, four month old male and female APP NL-F/NL-F mice were treated monthly with vehicle, 5 mg/kg Dasatinib + 50 mg/kg Quercetin, or 100 mg/kg Fisetin. Blood glucose levels, energy metabolism, spatial memory, amyloid burden, and senescent cell markers were assayed. Dasatinib + Quercetin treatment in female APP NL-F/NL-F mice increased oxygen consumption and energy expenditure resulting in decreased body mass. White adipose tissue mass was decreased along with senescence markers, SASP, blood glucose, and plasma insulin and triglycerides. Hippocampal senescence markers and SASP were reduced along with soluble and insoluble amyloid-β (Aβ) 42 and senescence associated-β-gal activity leading to improved spatial memory. Fisetin had negligible effects on these measures in female APP NL-F/NL-F mice while neither senolytic intervention altered these parameters in the male mice. Considering women have a greater risk of dementia, identifying senotherapeutics appropriate for sex and disease stage is necessary for personalized medicine.
Collapse
|
12
|
Garcia DN, Hense JD, Zanini BM, Isola JVV, Prosczek JB, Ashiqueali S, Oliveira TL, Mason JB, Schadock IC, Barros CC, Stout MB, Masternak MM, Schneider A. Senolytic treatment fails to improve ovarian reserve or fertility in female mice. GeroScience 2024; 46:3445-3455. [PMID: 38358579 PMCID: PMC11009191 DOI: 10.1007/s11357-024-01089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Senescent cell number increases with age in different tissues, leading to greater senescent cell load, proinflammatory stress, and tissue dysfunction. In the current study, we tested the efficacy of senolytic drugs to reduce ovarian senescence and improve fertility in reproductive age female mice. In the first experiment, 1-month-old C57BL/6 female mice were treated every other week with D + Q (n = 24) or placebo (n = 24). At 3 and 6 months of age, female mice were mated with untreated males to evaluate pregnancy rate and litter size. In the second experiment, 6-month-old C57BL/6 female mice were treated monthly with D + Q (n = 30), fisetin (n = 30), or placebo (n = 30). Females were treated once a month until 11 months of age, then they were mated with untreated males for 30 days to evaluate pregnancy rate and litter size. In the first experiment, D + Q treatment did not affect pregnancy rate (P = 0.68), litter size (P = 0.58), or ovarian reserve (P > 0.05). Lipofuscin staining was lower in females treated with D + Q (P = 0.04), but expression of senescence genes in ovaries was similar. In the second experiment, D + Q or fisetin treatment also did not affect pregnancy rate (P = 0.37), litter size (P = 0.20), or ovarian reserve (P > 0.05). Lipofuscin staining (P = 0.008) and macrophage infiltration (P = 0.002) was lower in fisetin treated females. Overall, treatment with D + Q or fisetin did not affect ovarian reserve or fertility but did decrease some senescence markers in the ovary.
Collapse
Affiliation(s)
- Driele N Garcia
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil
| | - Jessica D Hense
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil
| | - Bianka M Zanini
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil
| | - Jose V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Juliane B Prosczek
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil
| | - Sarah Ashiqueali
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Thais L Oliveira
- Centro de Biotecnologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeffrey B Mason
- College of Veterinary Medicine, Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, Utah State University, Logan, UT, USA
| | - Ines C Schadock
- Experimental Toxicology Department, German Institute for Risk Assessment, Berlin, Germany
| | - Carlos C Barros
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil
| | - Michael B Stout
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 - Sala 239, Pelotas, RS, CEP 96010-610, Brazil.
| |
Collapse
|
13
|
Bramwell LR, Frankum R, Harries LW. Repurposing Drugs for Senotherapeutic Effect: Potential Senomorphic Effects of Female Synthetic Hormones. Cells 2024; 13:517. [PMID: 38534362 DOI: 10.3390/cells13060517] [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: 01/26/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Repurposing previously approved drugs may fast track the route to the clinic for potential senotherapeutics and improves the inefficiency of the clinical drug development pipeline. We performed a repurposing screen of 240 clinically approved molecules in human primary dermal fibroblasts for their effects on CDKN2A expression. Molecules demonstrating effects on CDKN2A expression underwent secondary screening for senescence-associated beta galactosidase (SAB) activity, based on effect size, direction, and/or molecule identity. Selected molecules then underwent a more detailed assessment of senescence phenotypes including proliferation, apoptosis, DNA damage, senescence-associated secretory phenotype (SASP) expression, and regulators of alternative splicing. A selection of the molecules demonstrating effects on senescence were then used in a new bioinformatic structure-function screen to identify common structural motifs. In total, 90 molecules displayed altered CDKN2A expression at one or other dose, of which 15 also displayed effects on SAB positivity in primary human dermal fibroblasts. Of these, 3 were associated with increased SAB activity, and 11 with reduced activity. The female synthetic sex hormones-diethylstilboestrol, ethynyl estradiol and levonorgestrel-were all associated with a reduction in aspects of the senescence phenotype in male cells, with no effects visible in female cells. Finally, we identified that the 30 compounds that decreased CDKN2A activity the most had a common substructure linked to this function. Our results suggest that several drugs licensed for other indications may warrant exploration as future senotherapies, but that different donors and potentially different sexes may respond differently to senotherapeutic compounds. This underlines the importance of considering donor-related characteristics when designing drug screening platforms.
Collapse
Affiliation(s)
- Laura R Bramwell
- RNA-Mediated Mechanisms of Disease Group, Department of Clinical and Biomedical Sciences (Medical School), Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
| | - Ryan Frankum
- RNA-Mediated Mechanisms of Disease Group, Department of Clinical and Biomedical Sciences (Medical School), Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
| | - Lorna W Harries
- RNA-Mediated Mechanisms of Disease Group, Department of Clinical and Biomedical Sciences (Medical School), Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
| |
Collapse
|
14
|
Lee E, Carreras-Gallo N, Lopez L, Turner L, Lin A, Mendez TL, Went H, Tomusiak A, Verdin E, Corley M, Ndhlovu L, Smith R, Dwaraka VB. Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA methylation clocks: a longitudinal study on senolytic interventions. Aging (Albany NY) 2024; 16:3088-3106. [PMID: 38393697 PMCID: PMC10929829 DOI: 10.18632/aging.205581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
Senolytics, small molecules targeting cellular senescence, have emerged as potential therapeutics to enhance health span. However, their impact on epigenetic age remains unstudied. This study aimed to assess the effects of Dasatinib and Quercetin (DQ) senolytic treatment on DNA methylation (DNAm), epigenetic age, and immune cell subsets. In a Phase I pilot study, 19 participants received DQ for 6 months, with DNAm measured at baseline, 3 months, and 6 months. Significant increases in epigenetic age acceleration were observed in first-generation epigenetic clocks and mitotic clocks at 3 and 6 months, along with a notable decrease in telomere length. However, no significant differences were observed in second and third-generation clocks. Building upon these findings, a subsequent investigation evaluated the combination of DQ with Fisetin (DQF), a well-known antioxidant and antiaging senolytic molecule. After one year, 19 participants (including 10 from the initial study) received DQF for 6 months, with DNAm assessed at baseline and 6 months. Remarkably, the addition of Fisetin to the treatment resulted in non-significant increases in epigenetic age acceleration, suggesting a potential mitigating effect of Fisetin on the impact of DQ on epigenetic aging. Furthermore, our analyses unveiled notable differences in immune cell proportions between the DQ and DQF treatment groups, providing a biological basis for the divergent patterns observed in the evolution of epigenetic clocks. These findings warrant further research to validate and comprehensively understand the implications of these combined interventions.
Collapse
Affiliation(s)
- Edwin Lee
- Institute For Hormonal Balance, Orlando, FL 32819, USA
| | | | | | | | - Aaron Lin
- TruDiagnostic, Lexington, KY 40503, USA
| | | | | | - Alan Tomusiak
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | | | | |
Collapse
|
15
|
Lelarge V, Capelle R, Oger F, Mathieu T, Le Calvé B. Senolytics: from pharmacological inhibitors to immunotherapies, a promising future for patients' treatment. NPJ AGING 2024; 10:12. [PMID: 38321020 PMCID: PMC10847408 DOI: 10.1038/s41514-024-00138-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
The involvement of cellular senescence in the initiation and propagation of diseases is clearly characterized, making the elimination of senescent cells essential to treat age-related diseases. The development of senolytic drugs demonstrated that targeting these cells limits the deterioration of patients' condition, by inducing apoptosis. Nevertheless, the first generations of senolytics which has been developed displayed their activities through specific mechanisms and demonstrated several limitations during clinical development. However, the rational to eliminate senescent cells remains evident, with the necessity to develop specific therapies in a context of diseases and tissues. The evolutions in the field of drug discovery open the way to a new generation of senolytic therapies, such as immunological approaches (CAR-T cells, Antibody-Drug Conjugated or vaccines), which require preliminary steps of research to identify markers specifically expressed on senescent cells, demonstrating promising specific effects. Currently, the preclinical development of these strategies appears more challenging to avoid strong side effects, but the expected results are commensurate with patients' hopes for treatments. In this review, we highlight the fact that the classical senolytic approach based on drug repurposing display limited efficacy and probably reached its limits in term of clinical development. The recent development of more complex therapies and the extension of interest in the domain of senescence in different fields of research allow to extend the possibility to discover powerful therapies. The future of age-related diseases treatment is linked to the development of new approaches based on cell therapy or immunotherapy to offer the best treatment for patients.
Collapse
Affiliation(s)
- V Lelarge
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - R Capelle
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - F Oger
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1283 - UMR 8199 - EGID, 59000, Lille, France
| | - T Mathieu
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
- Synlab, 60/62 Rue d'Hauteville, 75010, Paris, France
| | - B Le Calvé
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France.
| |
Collapse
|
16
|
Cha J, Aguayo-Mazzucato C, Thompson PJ. Pancreatic β-cell senescence in diabetes: mechanisms, markers and therapies. Front Endocrinol (Lausanne) 2023; 14:1212716. [PMID: 37720527 PMCID: PMC10501801 DOI: 10.3389/fendo.2023.1212716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic β-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent β-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the β-cell, identification of molecular markers that distinguish senescent from non-senescent β-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of β-cell senescence, highlighting insights from mouse models as well as studies on human islets and β-cells. We identify markers that have been used to detect β-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in β-cells, heterogeneity of senescent β-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.
Collapse
Affiliation(s)
- Jeeyeon Cha
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Peter J. Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|