1
|
de Oliveira Silva T, Lunardon G, Lino CA, de Almeida Silva A, Zhang S, Irigoyen MCC, Lu YW, Mably JD, Barreto-Chaves MLM, Wang DZ, Diniz GP. Senescent cell depletion alleviates obesity-related metabolic and cardiac disorders. Mol Metab 2025; 91:102065. [PMID: 39557194 PMCID: PMC11636344 DOI: 10.1016/j.molmet.2024.102065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/06/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity is a major contributor to metabolic and cardiovascular disease. Although senescent cells have been shown to accumulate in adipose tissue, the role of senescence in obesity-induced metabolic disorders and in cardiac dysfunction is not yet clear; therefore, the therapeutic potential of managing senescence in obesity-related metabolic and cardiac disorders remains to be fully defined. OBJECTIVE We investigated the beneficial effects of a senolytic cocktail (dasatinib and quercetin) on senescence and its influence on obesity-related parameters. METHODS AND RESULTS We found that the increase in body weight and adiposity, glucose intolerance, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic disorders which were induced by an obesogenic diet were alleviated by senolytic cocktail treatment in mice. Treatment with senolytic compounds eliminated senescent cells, counteracting the activation of the senescence program and DNA damage in white adipose tissue (WAT) observed with an obesogenic diet. Moreover, the senolytic cocktail prevented the brown adipose tissue (BAT) whitening and increased the expression of the thermogenic gene profile in BAT and pWAT. In the hearts of obese mice, senolytic combination abolished myocardial maladaptation, reducing the senescence-associated secretory phenotype (SASP) and DNA damage, repressing cardiac hypertrophy, and improving diastolic dysfunction. Additionally, we showed that treatment with the senolytic cocktail corrected gene expression programs associated with fatty acid metabolism, oxidative phosphorylation, the P53 pathway, and DNA repair, which were all downregulated in obese mice. CONCLUSIONS Collectively, these data suggest that a senolytic cocktail can prevent the activation of the senescence program in the heart and WAT and activate the thermogenic program in BAT. Our results suggest that targeting senescent cells may be a novel therapeutic strategy for alleviating obesity-related metabolic and cardiac disorders.
Collapse
Affiliation(s)
- Tábatha de Oliveira Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Guilherme Lunardon
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Amanda de Almeida Silva
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Shiju Zhang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Yao Wei Lu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John D Mably
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Da-Zhi Wang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
2
|
Costa LNC, de Paula TP, Zazula MF, Naliwaiko K, Nassar CA, Bertolini GRF, Torrejais MM, Ribeiro LDFC, Costa RM. Maternal periodontitis potentiates monosodium glutamate-obesity damage on Wistar offspring's fast-glycolytic muscle. Oral Dis 2024; 30:4705-4720. [PMID: 38316639 DOI: 10.1111/odi.14890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/28/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024]
Abstract
OBJECTIVE To evaluate the effects of magnifying the damage caused by obesity induced by monosodium glutamate, using a model of maternal periodontitis, on the structure of the anterior tibialis muscle of the offspring. MATERIALS AND METHODS Twenty-four female Wistar rats were divided into four experimental groups: control (n = 6), obese (n = 6), control with periodontitis (n = 6) and obese with periodontitis (n = 6). At 78 days of life, the rats were mated with males without any experimental intervention. The offspring of these rats (n = 1/L), at 120 days of life, were weighed and measured, then euthanized. Plasma was collected for analysis of cytokines IL-6, IL-10, IL-17 and TNF-α. Adipose tissues were collected and weighed, and the anterior tibial muscle was designated for histomorphological analyses (n = 6/group). RESULTS Monosodium glutamate offspring showed significant muscle changes, such as a reduction in the size of fibres and neuromuscular junctions, and an increase in the nucleus and capillaries. However, all these changes were more expressed in monosodium glutamate-obese with periodontitis offspring. CONCLUSION This leads us to suggest a magnifying effect promoted by periodontitis to the damage already well described by monosodium glutamate-obesity, determined by low-intensity inflammation, causing greater muscle damage.
Collapse
Affiliation(s)
- Liziane Nunes Conrad Costa
- Laboratório de Biologia Estrutural e Funcional, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Thayná Petry de Paula
- Instituto de Pesquisa e Pós-Graduação em Odontologia, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Matheus Felipe Zazula
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Katya Naliwaiko
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Carlos Augusto Nassar
- Instituto de Pesquisa e Pós-Graduação em Odontologia, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Gladson Ricardo Flor Bertolini
- Laboratório de Estudo de Lesões e Recursos Fisioterapêuticos, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Marcia Miranda Torrejais
- Laboratório Experimental de Morfologia, Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Lucinéia de Fátima Chasko Ribeiro
- Laboratório de Biologia Estrutural e Funcional, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| | - Rose Meire Costa
- Laboratório de Biologia Estrutural e Funcional, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Paraná, Brazil
| |
Collapse
|
3
|
Fan S, Cai Y, Wei Y, Yang J, Gao J, Yang Y. Sarcopenic obesity and osteoporosis: Research progress and hot spots. Exp Gerontol 2024; 195:112544. [PMID: 39147076 DOI: 10.1016/j.exger.2024.112544] [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: 06/10/2024] [Revised: 07/17/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
Sarcopenic obesity (SO) and osteoporosis (OP) are associated with aging and obesity. The pathogenesis of SO is complex, including glucolipid and skeletal muscle metabolic disorders caused by inflammation, insulin resistance, and other factors. Growing evidence links muscle damage to bone loss. Muscle-lipid metabolism disorders of SO disrupt the balance between bone formation and bone resorption, increasing the risk of OP. Conversely, bones also play a role in fat and muscle metabolism. In the context of aging and obesity, the comprehensive review focuses on the effects of mechanical stimulation, mesenchymal stem cells (MSCs), chronic inflammation, myokines, and adipokines on musculoskeletal, at the same time, the impact of osteokines on muscle-lipid metabolism were also analyzed. So far, exercise combined with diet therapy is the most effective strategy for increasing musculoskeletal mass. A holistic treatment of musculoskeletal diseases is still in the preliminary exploration stage. Therefore, this article aims to improve the understanding of musculoskeletal -fat interactions in SO and OP, explores targets that can provide holistic treatment for SO combined with OP, and discusses current limitations and challenges. We hope to provide relevant ideas for developing specific therapies and improving disease prognosis in the future.
Collapse
Affiliation(s)
- Shangheng Fan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Yulan Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yunqin Wei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Department of Pharmacology, Zunyi Medical University, Zunyi, China.
| | - Yan Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
4
|
Lima MCDAM, Zazula MF, Martins LF, Carvalhal SR, Guimarães ATB, Fernandes LC, Naliwaiko K. How soon do metabolic alterations and oxidative distress precede the reduction of muscle mass and strength in Wistar rats in aging process? Biogerontology 2024; 25:491-506. [PMID: 38064115 DOI: 10.1007/s10522-023-10078-3] [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: 12/19/2022] [Accepted: 10/30/2023] [Indexed: 06/01/2024]
Abstract
Here we investigate metabolic changes, the antioxidant system and the accumulation of oxidative damage in muscles with different fiber types during the aging process in Wistar rats and try to map how sooner the changes occur. To do so, 30 male Wistar rats were submitted to behavioral evaluation to determine voluntary strength in the 11, 15, and 19 month old rats, measuring the energy metabolism, antioxidant system, oxidative damage and structure in the soleus and extensor digitorum longus muscles. We detected structural and metabolic changes in both muscles, especially in the EDL of 15 month old rats and in the soleus of 19 month old rats. In the 15 month old rats, there was a reduction in the cross-sectional area of the fibers, and a reduction in the proportion of type I fibers, accompanied by an increase in fiber density and the amount of type IIA fibers. This change in the fiber profile was followed by an increase in the activity of anaerobic metabolism enzymes, suggesting a reduction in the oxidative capacity of the muscle. In addition, there was an increase in the rate of lipid peroxidation, accompanied by a reduced antioxidant capacity. In the 19 month old rats, these disturbances got stronger. In summary, the present study demonstrated that before functional disturbances, there was an accumulation of oxidative damage and structural changes in the skeletal muscle beginning at 15 months old in the EDL and the soleus only in the biochemical parameters. Therefore, the metabolic alterations occurred at 15 months old and not before.
Collapse
Affiliation(s)
- Malu Cristina de Araújo Montoro Lima
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil
| | - Matheus Felipe Zazula
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil
| | - Luiz Fernando Martins
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil
| | - Stephanie Rubiane Carvalhal
- Laboratório de Metabolismo Celular, Departamento de Fisiologia, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil
| | - Ana Tereza Bittencourt Guimarães
- Laboratório de Investigações Biológicas, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Rua Universitária, 1619, Jardim Universitário, Cascavel, PR, 85819-110, Brazil
| | - Luiz Claudio Fernandes
- Laboratório de Metabolismo Celular, Departamento de Fisiologia, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil
| | - Katya Naliwaiko
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Setor de Ciências Biológicas, Universidade Federal do Paraná, Avenida Coronel Francisco Heráclito dos Santos, 100, Jardim das Américas, Curitiba, PR, 81530-900, Brazil.
| |
Collapse
|
5
|
Nolt GL, Keeble AR, Wen Y, Strong AC, Thomas NT, Valentino TR, Brightwell CR, Murach KA, Patrizia S, Weinstabl H, Gollner A, McCarthy JJ, Fry CS, Franti M, Filareto A, Peterson CA, Dungan CM. Inhibition of p53-MDM2 binding reduces senescent cell abundance and improves the adaptive responses of skeletal muscle from aged mice. GeroScience 2024; 46:2153-2176. [PMID: 37872294 PMCID: PMC10828311 DOI: 10.1007/s11357-023-00976-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Skeletal muscle adaptation to external stimuli, such as regeneration following injury and hypertrophy in response to resistance exercise, are blunted with advanced age. The accumulation of senescent cells, along with defects in myogenic progenitor cell (MPC) proliferation, have been strongly linked as contributing factors to age-associated impairment in muscle adaptation. p53 plays an integral role in all these processes, as upregulation of p53 causes apoptosis in senescent cells and prevents mitotic catastrophe in MPCs from old mice. The goal of this study was to determine if a novel pharmaceutical agent (BI01), which functions by upregulating p53 through inhibition of binding to MDM2, the primary p53 regulatory protein, improves muscle regeneration and hypertrophy in old mice. BI01 effectively reduced the number of senescent cells in vitro but had no effect on MPC survival or proliferation at a comparable dose. Following repeated oral gavage with 2 mg/kg of BI01 (OS) or vehicle (OV), old mice (24 months) underwent unilateral BaCl2 injury in the tibialis anterior (TA) muscle, with PBS injections serving as controls. After 7 days, satellite cell number was higher in the TA of OS compared to OV mice, as was the expression of genes involved in ATP production. By 35 days, old mice treated with BI01 displayed reduced senescent cell burden, enhanced regeneration (higher muscle mass and fiber cross-sectional area) and restoration of muscle function relative to OV mice. To examine the impact of 2 mg/kg BI01 on muscle hypertrophy, the plantaris muscle was subjected to 28 days of mechanical overload (MOV) in OS and OV mice. In response to MOV, OS mice had larger plantaris muscles and muscle fibers than OV mice, particularly type 2b + x fibers, associated with reduced senescent cells. Together our data show that BI01 is an effective senolytic agent that may also augment muscle metabolism to enhance muscle regeneration and hypertrophy in old mice.
Collapse
Affiliation(s)
- Georgia L Nolt
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Alexander R Keeble
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Yuan Wen
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Aubrey C Strong
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Nicholas T Thomas
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Taylor R Valentino
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Camille R Brightwell
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Sini Patrizia
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Harald Weinstabl
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - Andreas Gollner
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Michael Franti
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Antonio Filareto
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA.
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, Waco, TX, 76706, USA.
| |
Collapse
|
6
|
Gerosa L, Malvandi AM, Gomarasca M, Verdelli C, Sansoni V, Faraldi M, Ziemann E, Olivieri F, Banfi G, Lombardi G. Murine Myoblasts Exposed to SYUIQ-5 Acquire Senescence Phenotype and Differentiate into Sarcopenic-Like Myotubes, an In Vitro Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae022. [PMID: 38267369 PMCID: PMC10924451 DOI: 10.1093/gerona/glae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Indexed: 01/26/2024] Open
Abstract
The musculoskeletal system is one of the most affected organs by aging that correlates well with an accumulation of senescent cells as for other multiple age-related pathologies. The molecular mechanisms underpinning muscle impairment because of senescent cells are still elusive. The availability of in vitro model of skeletal muscle senescence is limited and restricted to a small panel of phenotypic features of these senescent cells in vivo. Here, we developed a new in vitro model of senescent C2C12 mouse myoblasts that, when subjected to differentiation, the resulting myotubes showed sarcopenic features. To induce senescence, we used SYUIQ-5, a quindoline derivative molecule inhibitor of telomerase activity, leading to the expression of several senescent hallmarks in treated myoblasts. They had increased levels of p21 protein accordingly with the observed cell cycle arrest. Furthermore, they had enhanced SA-βgalactosidase enzyme activity and phosphorylation of p53 and histone H2AX. SYUIQ-5 senescent myoblasts had impaired differentiation potential and the resulting myotubes showed increased levels of ATROGIN-1 and MURF1, ubiquitin ligases components responsible for protein degradation, and decreased mitochondria content, typical features of sarcopenic muscles. Myotubes differentiated from senescent myoblasts cultures release increased levels of MYOSTATIN that could affect skeletal muscle cell growth. Overall, our data suggest that a greater burden of senescent muscle cells could contribute to sarcopenia. This study presents a well-defined in vitro model of muscle cell senescence useful for deeper investigation in the aging research field to discover new putative therapeutic targets and senescence biomarkers associated with the aged musculoskeletal system.
Collapse
Affiliation(s)
- Laura Gerosa
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Marta Gomarasca
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Chiara Verdelli
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Veronica Sansoni
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Martina Faraldi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Fabiola Olivieri
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
7
|
Kamal M, Joanisse S, Parise G. Bleomycin-treated myoblasts undergo p21-associated cellular senescence and have severely impaired differentiation. GeroScience 2024; 46:1843-1859. [PMID: 37751045 PMCID: PMC10828175 DOI: 10.1007/s11357-023-00929-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
As we age, the ability to regenerate and repair skeletal muscle damage declines, partially due to increasing dysfunction of muscle resident stem cells-satellite cells (SC). Recent evidence implicates cellular senescence, which is the irreversible arrest of proliferation, as a potentiator of SC impairment during aging. However, little is known about the role of senescence in SC, and there is a large discrepancy in senescence classification within skeletal muscle. The purpose of this study was to develop a model of senescence in skeletal muscle myoblasts and identify how common senescence-associated biomarkers respond. Low-passage C2C12 myoblasts were treated with bleomycin or vehicle and then evaluated for cytological and molecular senescence markers, proliferation status, cell cycle kinetics, and differentiation potential. Bleomycin treatment caused double-stranded DNA breaks, which upregulated p21 mRNA and protein, potentially through NF-κB and senescence-associated super enhancer (SASE) signaling (p < 0.01). Consequently, cell proliferation was abruptly halted due to G2/M-phase arrest (p < 0.01). Bleomycin-treated myoblasts displayed greater senescence-associated β-galactosidase staining (p < 0.01), which increased over several days. These myoblasts remained senescent following 6 days of differentiation and had significant impairments in myotube formation (p < 0.01). Furthermore, our results show that senescence can be maintained despite the lack of p16 gene expression in C2C12 myoblasts. In conclusion, bleomycin treatment provides a valid model of damage-induced senescence that was associated with elevated p21, reduced myoblast proliferation, and aberrant cell cycle kinetics, while confirming that a multi-marker approach is needed for the accurate classification of senescence within skeletal muscle.
Collapse
Affiliation(s)
- Michael Kamal
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Sophie Joanisse
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
- Department of Sport and Exercise Sciences, Musculoskeletal Science and Sport Medicine Research Centre, Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Gianni Parise
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
8
|
Habiballa L, Hruby A, Granic A, Dodds RM, Hillman SJ, Jurk D, Passos JF, Sayer AA. Determining the feasibility of characterising cellular senescence in human skeletal muscle and exploring associations with muscle morphology and physical function at different ages: findings from the MASS_Lifecourse Study. GeroScience 2024; 46:1141-1158. [PMID: 37434081 PMCID: PMC10828484 DOI: 10.1007/s11357-023-00869-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023] Open
Abstract
Cellular senescence may be associated with morphological changes in skeletal muscle and changes in physical function with age although there have been few human studies. We aimed to determine the feasibility of characterising cellular senescence in skeletal muscle and explored sex-specific associations between markers of cellular senescence, muscle morphology, and physical function in participants from the MASS_Lifecourse Study. Senescence markers (p16, TAF (Telomere-Associated DNA Damage Foci), HMGB1 (High Mobility Group Box 1), and Lamin B1) and morphological characteristics (fibre size, number, fibrosis, and centrally nucleated fibres) were assessed in muscle biopsies from 40 men and women (age range 47-84) using spatially-resolved methods (immunohistochemistry, immunofluorescence, and RNA and fluorescence in situ hybridisation). The associations between senescence, morphology, and physical function (muscle strength, mass, and physical performance) at different ages were explored. We found that most senescence markers and morphological characteristics were weakly associated with age in men but more strongly, although non-significantly, associated with age in women. Associations between senescence markers, morphology, and physical function were also stronger in women for HMGB1 and grip strength (r = 0.52); TAF, BMI, and muscle mass (r > 0.4); Lamin B1 and fibrosis (r = - 0.5); fibre size and muscle mass (r ≥ 0.4); and gait speed (r = - 0.5). However, these associations were non-significant. In conclusion, we have demonstrated that it is feasible to characterise cellular senescence in human skeletal muscle and to explore associations with morphology and physical function in women and men of different ages. The findings require replication in larger studies.
Collapse
Affiliation(s)
- Leena Habiballa
- AGE Research Group, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Adam Hruby
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- University of Southern California, Los Angeles, CA, USA
| | - Antoneta Granic
- AGE Research Group, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.
| | - Richard M Dodds
- AGE Research Group, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Susan J Hillman
- AGE Research Group, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Diana Jurk
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - João F Passos
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Avan A Sayer
- AGE Research Group, Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
9
|
Ismaeel A, McDermott MM, Joshi JK, Sturgis JC, Zhang D, Ho KJ, Sufit R, Ferrucci L, Peterson CA, Kosmac K. Cocoa flavanols, Nrf2 activation, and oxidative stress in peripheral artery disease: mechanistic findings in muscle based on outcomes from a randomized trial. Am J Physiol Cell Physiol 2024; 326:C589-C605. [PMID: 38189132 PMCID: PMC11193455 DOI: 10.1152/ajpcell.00573.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/09/2024]
Abstract
The pathophysiology of muscle damage in peripheral artery disease (PAD) includes increased oxidant production and impaired antioxidant defenses. Epicatechin (EPI), a naturally occurring flavanol, has antioxidant properties that may mediate the beneficial effects of natural products such as cocoa. In a phase II randomized trial, a cocoa-flavanol-rich beverage significantly improved walking performance compared with a placebo in people with PAD. In the present work, the molecular mechanisms underlying the therapeutic effect of cocoa flavanols were investigated by analyzing baseline and follow-up muscle biopsies from participants. Increases in nuclear factor erythroid 2-related factor 2 (Nrf2) target antioxidants heme oxygenase-1 (HO-1) and NAD(P)H dehydrogenase [quinone] 1 (NQO1) in the cocoa group were significantly associated with reduced accumulation of central nuclei, a myopathy indicator, in type II muscle fibers (P = 0.017 and P = 0.023, respectively). Protein levels of the mitochondrial respiratory complex III subunit, cytochrome b-c1 complex subunit 2 (UQCRC2), were significantly higher in the cocoa group than in the placebo group (P = 0.032), and increases in UQCRC2 were significantly associated with increased levels of Nrf2 target antioxidants HO-1 and NQO1 (P = 0.001 and P = 0.035, respectively). Exposure of non-PAD human myotubes to ex vivo serum from patients with PAD reduced Nrf2 phosphorylation, an indicator of activation, increased hydrogen peroxide production and oxidative stress, and reduced mitochondrial respiration. Treatment of myotubes with EPI in the presence of serum from patients with PAD increased Nrf2 phosphorylation and protected against PAD serum-induced oxidative stress and mitochondrial dysfunction. Overall, these findings suggest that cocoa flavanols may enhance antioxidant capacity in PAD via Nrf2 activation.NEW & NOTEWORTHY The current study supports the hypothesis that in people with PAD, cocoa flavanols activate Nrf2, thereby increasing antioxidant protein levels, protecting against skeletal muscle damage, and increasing mitochondrial protein abundance. These results suggest that Nrf2 activation may be an important therapeutic target for improving walking performance in people with PAD.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Deparment of Physiology, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - Mary M McDermott
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Jai K Joshi
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky, United States
| | - Jada C Sturgis
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky, United States
| | - Dongxue Zhang
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Karen J Ho
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Robert Sufit
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Luigi Ferrucci
- National Institute on Aging, Intramural Research Program, Baltimore, Maryland, United States
| | - Charlotte A Peterson
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky, United States
| | - Kate Kosmac
- Department of Physical Therapy, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
10
|
Rex N, Melk A, Schmitt R. Cellular senescence and kidney aging. Clin Sci (Lond) 2023; 137:1805-1821. [PMID: 38126209 PMCID: PMC10739085 DOI: 10.1042/cs20230140] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Life expectancy is increasing worldwide, and by 2050 the proportion of the world's population over 65 years of age is estimated to surpass 1.5 billion. Kidney aging is associated with molecular and physiological changes that cause a loss of renal function and of regenerative potential. As the aging population grows, it is crucial to understand the mechanisms underlying these changes, as they increase the susceptibility to developing acute kidney injury (AKI) and chronic kidney disease (CKD). Various cellular processes and molecular pathways take part in the complex process of kidney aging. In this review, we will focus on the phenomenon of cellular senescence as one of the involved mechanisms at the crossroad of kidney aging, age-related disease, and CKD. We will highlight experimental and clinical findings about the role of cellular senescence in kidney aging and CKD. In addition, we will review challenges in senescence research and emerging therapeutic aspects. We will highlight the great potential of senolytic strategies for the elimination of harmful senescent cells to promote healthy kidney aging and to avoid age-related disease and CKD. This review aims to give insight into recent discoveries and future developments, providing a comprehensive overview of current knowledge on cellular senescence and anti-senescent therapies in the kidney field.
Collapse
Affiliation(s)
- Nikolai Rex
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Germany
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
11
|
Terrell K, Choi S, Choi S. Calcium's Role and Signaling in Aging Muscle, Cellular Senescence, and Mineral Interactions. Int J Mol Sci 2023; 24:17034. [PMID: 38069357 PMCID: PMC10706910 DOI: 10.3390/ijms242317034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Calcium research, since its pivotal discovery in the early 1800s through the heating of limestone, has led to the identification of its multi-functional roles. These include its functions as a reducing agent in chemical processes, structural properties in shells and bones, and significant role in cells relating to this review: cellular signaling. Calcium signaling involves the movement of calcium ions within or between cells, which can affect the electrochemical gradients between intra- and extracellular membranes, ligand binding, enzyme activity, and other mechanisms that determine cell fate. Calcium signaling in muscle, as elucidated by the sliding filament model, plays a significant role in muscle contraction. However, as organisms age, alterations occur within muscle tissue. These changes include sarcopenia, loss of neuromuscular junctions, and changes in mineral concentration, all of which have implications for calcium's role. Additionally, a field of study that has gained recent attention, cellular senescence, is associated with aging and disturbed calcium homeostasis, and is thought to affect sarcopenia progression. Changes seen in calcium upon aging may also be influenced by its crosstalk with other minerals such as iron and zinc. This review investigates the role of calcium signaling in aging muscle and cellular senescence. We also aim to elucidate the interactions among calcium, iron, and zinc across various cells and conditions, ultimately deepening our understanding of calcium signaling in muscle aging.
Collapse
Affiliation(s)
| | | | - Sangyong Choi
- Department of Nutritional Sciences, College of Agriculture, Health, and Natural Resources, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
12
|
Granic A, Suetterlin K, Shavlakadze T, Grounds M, Sayer A. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men. Clin Sci (Lond) 2023; 137:1721-1751. [PMID: 37986616 PMCID: PMC10665130 DOI: 10.1042/cs20230319] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Ageing is a complex biological process associated with increased morbidity and mortality. Nine classic, interdependent hallmarks of ageing have been proposed involving genetic and biochemical pathways that collectively influence ageing trajectories and susceptibility to pathology in humans. Ageing skeletal muscle undergoes profound morphological and physiological changes associated with loss of strength, mass, and function, a condition known as sarcopenia. The aetiology of sarcopenia is complex and whilst research in this area is growing rapidly, there is a relative paucity of human studies, particularly in older women. Here, we evaluate how the nine classic hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication contribute to skeletal muscle ageing and the pathophysiology of sarcopenia. We also highlight five novel hallmarks of particular significance to skeletal muscle ageing: inflammation, neural dysfunction, extracellular matrix dysfunction, reduced vascular perfusion, and ionic dyshomeostasis, and discuss how the classic and novel hallmarks are interconnected. Their clinical relevance and translational potential are also considered.
Collapse
Affiliation(s)
- Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| | - Karen Suetterlin
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne, U.K
| | - Tea Shavlakadze
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, NY, U.S.A
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Avan A. Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| |
Collapse
|
13
|
Axelrod CL, Dantas WS, Kirwan JP. Sarcopenic obesity: emerging mechanisms and therapeutic potential. Metabolism 2023; 146:155639. [PMID: 37380015 PMCID: PMC11448314 DOI: 10.1016/j.metabol.2023.155639] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/08/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Sarcopenic obesity, or the loss of muscle mass and function associated with excess adiposity, is a largely untreatable medical condition associated with diminished quality of life and increased risk of mortality. To date, it remains somewhat paradoxical and mechanistically undefined as to why a subset of adults with obesity develop muscular decline, an anabolic stimulus generally associated with retention of lean mass. Here, we review evidence surrounding the definition, etiology, and treatment of sarcopenic obesity with an emphasis on emerging regulatory nodes with therapeutic potential. We review the available clinical evidence largely focused on diet, lifestyle, and behavioral interventions to improve quality of life in patients with sarcopenic obesity. Based upon available evidence, relieving consequences of energy burden, such as oxidative stress, myosteatosis, and/or mitochondrial dysfunction, is a promising area for therapeutic development in the treatment and management of sarcopenic obesity.
Collapse
Affiliation(s)
- Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
14
|
Sunnerberg J, Thomas WS, Petusseau A, Reed MS, Jack Hoopes P, Pogue BW. Review of optical reporters of radiation effects in vivo: tools to quantify improvements in radiation delivery technique. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:080901. [PMID: 37560327 PMCID: PMC10409499 DOI: 10.1117/1.jbo.28.8.080901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
Significance Radiation damage studies are used to optimize radiotherapy treatment techniques. Although biological indicators of damage are the best assays of effect, they are highly variable due to biological heterogeneity. The free radical radiochemistry can be assayed with optical reporters, allowing for high precision titration of techniques. Aim We examine the optical reporters of radiochemistry to highlight those with the best potential for translational use in vivo, as surrogates for biological damage assays, to inform on mechanisms. Approach A survey of the radical chemistry effects from reactive oxygen species (ROS) and oxygen itself was completed to link to DNA or biological damage. Optical reporters of ROS include fluorescent, phosphorescent, and bioluminescent molecules that have a variety of activation pathways, and each was reviewed for its in vivo translation potential. Results There are molecular reporters of ROS having potential to report within living systems, including derivatives of luminol, 2'7'-dichlorofluorescein diacetate, Amplex Red, and fluorescein. None have unique specificity to singular ROS species. Macromolecular engineered reporters unique to specific ROS are emerging. The ability to directly measure oxygen via reporters, such as Oxyphor and protoporphyrin IX, is an opportunity to quantify the consumption of oxygen during ROS generation, and this translates from in vitro to in vivo use. Emerging techniques, such as ion particle beams, spatial fractionation, and ultra-high dose rate FLASH radiotherapy, provide the motivation for these studies. Conclusions In vivo optical reporters of radiochemistry are quantitatively useful for comparing radiotherapy techniques, although their use comes at the cost of the unknown connection to the mechanisms of radiobiological damage. Still their lower measurement uncertainty, compared with biological response assay, makes them an invaluable tool. Linkage to DNA damage and biological damage is needed, and measures such as oxygen consumption serve as useful surrogate measures that translate to in vivo use.
Collapse
Affiliation(s)
- Jacob Sunnerberg
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - William S. Thomas
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Arthur Petusseau
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Matthew S. Reed
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - P. Jack Hoopes
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Brian W. Pogue
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| |
Collapse
|
15
|
Dungan CM, Wells JM, Murach KA. The life and times of cellular senescence in skeletal muscle: friend or foe for homeostasis and adaptation? Am J Physiol Cell Physiol 2023; 325:C324-C331. [PMID: 37335024 PMCID: PMC10393344 DOI: 10.1152/ajpcell.00553.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
A gradual decline in skeletal muscle mass and function is closely tied to increased mortality and disease risk during organismal aging. Exercise training is the most effective way to enhance muscle health, but the adaptive response to exercise as well as muscle repair potential is blunted in older individuals. Numerous mechanisms contribute to the loss of muscle mass and plasticity as aging progresses. An emerging body of recent evidence implicates an accumulation of senescent ("zombie") cells in muscle as a contributing factor to the aging phenotype. Senescent cells cannot divide but can release inflammatory factors and create an unfavorable environment for homeostasis and adaptation. On balance, some evidence indicates that cells with senescent characteristics can be beneficial for the muscle adaptive process, specifically at younger ages. Emerging evidence also suggests that multinuclear muscle fibers could become senescent. In this review, we summarize current literature on the prevalence of senescent cells in skeletal muscle and highlight the consequences of senescent cell removal on muscle mass, function, and adaptability. We examine key limitations in the field of senescence specifically in skeletal muscle and identify areas of research that require future investigation.NEW & NOTEWORTHY There is evidence to suggest that senescent "zombie" cells may or may not accrue in aging skeletal muscle. When muscle is perturbed regardless of age, senescent-like cells do appear, and the benefits of removing them could be age-dependent. More work is needed to determine the magnitude of accumulation and source of senescent cells in muscle. Regardless, pharmacological senolytic treatment of aged muscle is beneficial for adaptation.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, Texas, United States
| | - Jaden M Wells
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, Texas, United States
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
16
|
Witham MD, Granic A, Miwa S, Passos JF, Richardson GD, Sayer AA. New Horizons in cellular senescence for clinicians. Age Ageing 2023; 52:afad127. [PMID: 37466640 PMCID: PMC10355181 DOI: 10.1093/ageing/afad127] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Indexed: 07/20/2023] Open
Abstract
Cellular senescence has emerged as a fundamental biological mechanism underpinning the ageing process and has been implicated in the pathogenesis of an increasing number of age-related conditions. Cellular senescence is a cell fate originally defined as an irreversible loss of replicative potential although it is now clear that it can be induced by a variety of mechanisms independent of replication and telomere attrition. The drivers include a persistent DNA damage response causing multiple alterations in cellular function. Senescent cells secrete a range of mediators that drive chronic inflammation and can convert other cells to the senescent state-the senescence-associated secretory phenotype. Much research to date has been conducted in animal models, but it is now clear that senescent cells accompany ageing in humans and their presence is an important driver of disease across systems. Proof-of-concept work suggests that preventing or reversing senescence may be a viable strategy to counteract human ageing and age-related disease. Possible interventions include exercise, nutrition and senolytics/senostatic drugs although there are a number of potential limitations to the use of senotherapeutics. These interventions are generally tested for single-organ conditions, but the real power of this approach is the potential to tackle multiple age-related conditions. The litmus test for this exciting new class of therapies, however, will be whether they can improve healthy life expectancy rather than merely extending lifespan. The outcomes measured in clinical studies need to reflect these aims if senotherapeutics are to gain the trust of clinicians, patients and the public.
Collapse
Affiliation(s)
- Miles D Witham
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| | - Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Joao F Passos
- Department of Physiology and Biomedical Engineering and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Gavin D Richardson
- Vascular Medicine and Biology Theme, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Avan A Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| |
Collapse
|
17
|
A cross-talk between sestrins, chronic inflammation and cellular senescence governs the development of age-associated sarcopenia and obesity. Ageing Res Rev 2023; 86:101852. [PMID: 36642190 DOI: 10.1016/j.arr.2023.101852] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The rapid increase in both the lifespan and proportion of older adults is accompanied by the unprecedented rise in age-associated chronic diseases, including sarcopenia and obesity. Aging is also manifested by increased susceptibility to multiple endogenous and exogenous stresses enabling such chronic conditions to develop. Among the main physiological regulators of cellular adaption to various stress stimuli, such as DNA damage, hypoxia, and oxidative stress, are sestrins (Sesns), a family of three evolutionarily conserved proteins, Sesn1, 2, and 3. Age-associated sarcopenia and obesity are characterized by two key processes: (i) accumulation of senescent cells in the skeletal muscle and adipose tissue and (ii) creation of a systemic, chronic, low-grade inflammation (SCLGI). Presumably, failed SCLGI resolution governs the development of these chronic conditions. Noteworthy, Sesns activate senolytics, which are agents that selectively eliminate senescent cells, as well as specialized pro-resolving mediators, which are factors that physiologically provide inflammation resolution. Sesns reveal clear beneficial effects in pre-clinical models of sarcopenia and obesity. Based on these observations, we propose a novel treatment strategy for age-associated sarcopenia and obesity, complementary to the conventional therapeutic modalities: Sesn activation, SCLGI resolution, and senescent cell elimination.
Collapse
|
18
|
Mankhong S, Kim S, Moon S, Lee JS, Cho EJ, Kwak HB, Park DH, Ryu JK, Kang JH. Melatonin and Exercise Counteract Sarcopenic Obesity through Preservation of Satellite Cell Function. Int J Mol Sci 2023; 24:ijms24076097. [PMID: 37047070 PMCID: PMC10094434 DOI: 10.3390/ijms24076097] [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: 02/06/2023] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Sarcopenic obesity (SO) is characterized by atrophic skeletal muscle impairment (sarcopenia) and obesity, which is associated with adverse outcomes of morbidity and mortality in elderly people. We investigated the effects of melatonin and exercise training on SO in 32-week-old senescence-accelerated mouse-prone-8 (SAMP8) mice fed a normal diet or a high-fat diet for 16 weeks. Melatonin, exercise, or melatonin and exercise for 8 weeks displayed reductions in the SO-induced impairment of skeletal muscle function and atrophy. Specifically, a decrease in mitochondrial calcium retention capacity in skeletal muscles observed in the HFD-con group was attenuated in melatonin and/or exercise intervention groups. More importantly, HFD-con mice displayed a lower number of Pax7+ satellite cells (SCs) and higher expression of p16ink than P8ND mice, which were attenuated by melatonin and/or exercise interventions. The cellular senescence in SC-derived primary myoblasts from HFD-con mice was significantly attenuated in myoblasts from the melatonin and/or exercise groups, which was reproduced in a senescence model of H2O2-treated C2C12 myoblasts. Our results suggest that melatonin and exercise training attenuate SO-induced skeletal muscle dysfunction, at least in part, through preserving the SC pool by inhibiting cellular senescence and attenuating mitochondrial dysfunction.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Sujin Kim
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Jae-Seon Lee
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Eun-Jeong Cho
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Ji-Kan Ryu
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Urology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
19
|
Petrocelli JJ, de Hart NM, Lang MJ, Yee EM, Ferrara PJ, Fix DK, Chaix A, Funai K, Drummond MJ. Cellular senescence and disrupted proteostasis induced by myotube atrophy are prevented with low-dose metformin and leucine cocktail. Aging (Albany NY) 2023; 15:1808-1832. [PMID: 36947713 PMCID: PMC10085594 DOI: 10.18632/aging.204600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/24/2023]
Abstract
Aging coincides with the accumulation of senescent cells within skeletal muscle that produce inflammatory products, known as the senescence-associated secretory phenotype, but the relationship of senescent cells to muscle atrophy is unclear. Previously, we found that a metformin + leucine (MET+LEU) treatment had synergistic effects in aged mice to improve skeletal muscle structure and function during disuse atrophy. Therefore, the study's purpose was to determine the mechanisms by which MET+LEU exhibits muscle atrophy protection in vitro and if this occurs through cellular senescence. C2C12 myoblasts differentiated into myotubes were used to determine MET+LEU mechanisms during atrophy. Additionally, aged mouse single myofibers and older human donor primary myoblasts were individually isolated to determine the translational potential of MET+LEU on muscle cells. MET+LEU (25 + 125 μM) treatment increased myotube differentiation and prevented myotube atrophy. Low concentration (0.1 + 0.5 μM) MET+LEU had unique effects to prevent muscle atrophy and increase transcripts related to protein synthesis and decrease transcripts related to protein breakdown. Myotube atrophy resulted in dysregulated proteostasis that was reversed with MET+LEU and individually with proteasome inhibition (MG-132). Inflammatory and cellular senescence transcriptional pathways and respective transcripts were increased following myotube atrophy yet reversed with MET+LEU treatment. Dasatinib + quercetin (D+Q) senolytic prevented myotube atrophy similar to MET+LEU. Finally, MET+LEU prevented loss in myotube size in alternate in vitro models of muscle atrophy as well as in aged myofibers while, in human primary myotubes, MET+LEU prevented reductions in myonuclei fusion. These data support that MET+LEU has skeletal muscle cell-autonomous properties to prevent atrophy by reversing senescence and improving proteostasis.
Collapse
Affiliation(s)
- Jonathan J. Petrocelli
- Department of Physical Therapy and Athletic, University of Utah, Salt Lake City, UT 84112, USA
| | - Naomi M.M.P. de Hart
- Department of Physical Therapy and Athletic, University of Utah, Salt Lake City, UT 84112, USA
| | - Marisa J. Lang
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Elena M. Yee
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Patrick J. Ferrara
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Dennis K. Fix
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Katsuhiko Funai
- Department of Physical Therapy and Athletic, University of Utah, Salt Lake City, UT 84112, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Micah J. Drummond
- Department of Physical Therapy and Athletic, University of Utah, Salt Lake City, UT 84112, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
20
|
Lino CA, de Oliveira-Silva T, Lunardon G, Balbino-Silva C, Lima VM, Huang ZP, Donato J, Takano APC, Barreto-Chaves ML, Wang DZ, Diniz GP. Ablation of miRNA-22 protects against obesity-induced adipocyte senescence and ameliorates metabolic disorders in middle-aged mice. Mech Ageing Dev 2023; 210:111775. [PMID: 36641038 DOI: 10.1016/j.mad.2023.111775] [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: 10/04/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
High-fat diet (HFD) promotes obesity-related metabolic complications by activating cellular senescence in white adipose tissue (WAT). Growing evidence supports the importance of microRNA-22 (miR-22) in metabolic disorders and cellular senescence. Recently, we showed that miR-22 deletion attenuates obesity-related metabolic abnormalities. However, whether miR-22 mediates HFD-induced cellular senescence of WAT remains unknown. Here, we uncovered that obese mice displayed increased pri-miR-22 levels and cellular senescence in WAT. However, miR-22 ablation protected mice against HFD-induced WAT senescence. In addition, in vitro studies showed that miR-22 deletion prevented preadipocyte senescence in response to Doxorubicin (Doxo). Loss-of-function studies in vitro and in vivo revealed that miR-22 increases H2ax mRNA and γH2ax levels in preadipocytes and WAT without inducing DNA damage. Intriguingly, miR-22 ablation prevented HFD-induced increase in γH2ax levels and DNA damage in WAT. Similarly, miR-22 deletion prevented Doxo-induced increase in γH2ax levels in preadipocytes. Adipose miR-22 levels were enhanced in middle-aged mice fed a HFD than those found in young mice. Furthermore, miR-22 deletion attenuated fat mass gain and glucose imbalance induced by HFD in middle-aged mice. Overall, our findings indicate that miR-22 is a key regulator of obesity-induced WAT senescence and metabolic disorders in middle-aged mice.
Collapse
Affiliation(s)
- Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Guilherme Lunardon
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camila Balbino-Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vanessa M Lima
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Zhan-Peng Huang
- Center for Translational Medicine, The First Affiliated Hospital, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Paula C Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
21
|
Taglietti V, Kefi K, Rivera L, Bergiers O, Cardone N, Coulpier F, Gioftsidi S, Drayton-Libotte B, Hou C, Authier FJ, Pietri-Rouxel F, Robert M, Bremond-Gignac D, Bruno C, Fiorillo C, Malfatti E, Lafuste P, Tiret L, Relaix F. Thyroid-stimulating hormone receptor signaling restores skeletal muscle stem cell regeneration in rats with muscular dystrophy. Sci Transl Med 2023; 15:eadd5275. [PMID: 36857434 DOI: 10.1126/scitranslmed.add5275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe and progressive myopathy leading to motor and cardiorespiratory impairment. We analyzed samples from patients with DMD and a preclinical rat model of severe DMD and determined that compromised repair capacity of muscle stem cells in DMD is associated with early and progressive muscle stem cell senescence. We also found that extraocular muscles (EOMs), which are spared by the disease in patients, contain muscle stem cells with long-lasting regenerative potential. Using single-cell transcriptomics analysis of muscles from a rat model of DMD, we identified the gene encoding thyroid-stimulating hormone receptor (Tshr) as highly expressed in EOM stem cells. Further, TSHR activity was involved in preventing senescence. Forskolin, which activates signaling downstream of TSHR, was found to reduce senescence of skeletal muscle stem cells, increase stem cell regenerative potential, and promote myogenesis, thereby improving muscle function in DMD rats. These findings indicate that stimulation of adenylyl cyclase leads to muscle repair in DMD, potentially providing a therapeutic approach for patients with the disease.
Collapse
Affiliation(s)
| | - Kaouthar Kefi
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - Lea Rivera
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - Oriane Bergiers
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - Nastasia Cardone
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - Fanny Coulpier
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | | | | | - Cyrielle Hou
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - François-Jérôme Authier
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France.,AP-HP, Hôpital Mondor, FHU SENEC, Service d'histologie, F-94010 Créteil, France
| | - France Pietri-Rouxel
- Sorbonne Université, INSERM, UMRS974, Center for Research in Myology, F-75013, Paris, France
| | - Matthieu Robert
- Borelli centre, UMR 9010, CNRS - SSA - ENS Paris Saclay - Université Paris Cité, F-75016, Paris, France.,Ophthalmology Department, Necker Enfants Malades University Hospital, AP-HP, F-75015, Paris, France
| | - Dominique Bremond-Gignac
- Ophthalmology Department, Necker Enfants Malades University Hospital, AP-HP, F-75015, Paris, France.,INSERM, UMRS1138, Team 17, Sorbonne Paris Cité University, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Gaslini Institute, DINOGMI, University of Genova, 16147, Genova, Italy
| | - Chiara Fiorillo
- Center of Translational and Experimental Myology, IRCCS Gaslini Institute, DINOGMI, University of Genova, 16147, Genova, Italy
| | - Edoardo Malfatti
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France.,AP-HP, Hôpital Mondor, FHU SENEC, Service d'histologie, F-94010 Créteil, France
| | - Peggy Lafuste
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France
| | - Laurent Tiret
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France.,École nationale vétérinaire d'Alfort, IMRB, F-94700, Maisons-Alfort, France
| | - Frédéric Relaix
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010 Créteil, France.,AP-HP, Hôpital Mondor, FHU SENEC, Service d'histologie, F-94010 Créteil, France.,École nationale vétérinaire d'Alfort, IMRB, F-94700, Maisons-Alfort, France.,EFS, IMRB, F-94010, Creteil, France
| |
Collapse
|
22
|
An Early and Sustained Inflammatory State Induces Muscle Changes and Establishes Obesogenic Characteristics in Wistar Rats Exposed to the MSG-Induced Obesity Model. Int J Mol Sci 2023; 24:ijms24054730. [PMID: 36902158 PMCID: PMC10003260 DOI: 10.3390/ijms24054730] [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: 10/21/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 03/05/2023] Open
Abstract
The model of obesity induced by monosodium glutamate cytotoxicity on the hypothalamic nuclei is widely used in the literature. However, MSG promotes persistent muscle changes and there is a significant lack of studies that seek to elucidate the mechanisms by which damage refractory to reversal is established. This study aimed to investigate the early and chronic effects of MSG induction of obesity upon systemic and muscular parameters of Wistar rats. The animals were exposed to MSG subcutaneously (4 mg·g-1 b.w.) or saline (1.25 mg·g-1 b.w.) daily from PND01 to PND05 (n = 24). Afterwards, in PND15, 12 animals were euthanized to determine the plasma and inflammatory profile and to assess muscle damage. In PND142, the remaining animals were euthanized, and samples for histological and biochemical analyses were obtained. Our results suggest that early exposure to MSG reduced growth, increased adiposity, and inducted hyperinsulinemia and a pro-inflammatory scenario. In adulthood, the following were observed: peripheral insulin resistance, increased fibrosis, oxidative distress, and a reduction in muscle mass, oxidative capacity, and neuromuscular junctions, increased fibrosis, and oxidative distress. Thus, we can conclude that the condition found in adult life and the difficulty restoring in the muscle profile is related to the metabolic damage established early on.
Collapse
|
23
|
Bhardwaj P, Iyengar NM, Zahid H, Carter KM, Byun DJ, Choi MH, Sun Q, Savenkov O, Louka C, Liu C, Piloco P, Acosta M, Bareja R, Elemento O, Foronda M, Dow LE, Oshchepkova S, Giri DD, Pollak M, Zhou XK, Hopkins BD, Laughney AM, Frey MK, Ellenson LH, Morrow M, Spector JA, Cantley LC, Brown KA. Obesity promotes breast epithelium DNA damage in women carrying a germline mutation in BRCA1 or BRCA2. Sci Transl Med 2023; 15:eade1857. [PMID: 36812344 PMCID: PMC10557057 DOI: 10.1126/scitranslmed.ade1857] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023]
Abstract
Obesity, defined as a body mass index (BMI) ≥ 30, is an established risk factor for breast cancer among women in the general population after menopause. Whether elevated BMI is a risk factor for women with a germline mutation in BRCA1 or BRCA2 is less clear because of inconsistent findings from epidemiological studies and a lack of mechanistic studies in this population. Here, we show that DNA damage in normal breast epithelia of women carrying a BRCA mutation is positively correlated with BMI and with biomarkers of metabolic dysfunction. In addition, RNA sequencing showed obesity-associated alterations to the breast adipose microenvironment of BRCA mutation carriers, including activation of estrogen biosynthesis, which affected neighboring breast epithelial cells. In breast tissue explants cultured from women carrying a BRCA mutation, we found that blockade of estrogen biosynthesis or estrogen receptor activity decreased DNA damage. Additional obesity-associated factors, including leptin and insulin, increased DNA damage in human BRCA heterozygous epithelial cells, and inhibiting the signaling of these factors with a leptin-neutralizing antibody or PI3K inhibitor, respectively, decreased DNA damage. Furthermore, we show that increased adiposity was associated with mammary gland DNA damage and increased penetrance of mammary tumors in Brca1+/- mice. Overall, our results provide mechanistic evidence in support of a link between elevated BMI and breast cancer development in BRCA mutation carriers. This suggests that maintaining a lower body weight or pharmacologically targeting estrogen or metabolic dysfunction may reduce the risk of breast cancer in this population.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Heba Zahid
- Department of Medical Laboratory Technology, College of Applied Medical Science, Taibah University, Medina 42353, Saudi Arabia
| | | | - Dong Jun Byun
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Qi Sun
- Computational Biology Service Unit of Life Sciences Core Laboratories Center, Cornell University, Ithaca, NY 14853, USA
| | - Oleksandr Savenkov
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Charalambia Louka
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Catherine Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Phoebe Piloco
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Monica Acosta
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miguel Foronda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lukas E. Dow
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sofya Oshchepkova
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dilip D. Giri
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, Canada
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Benjamin D. Hopkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashley M. Laughney
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melissa K. Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lora Hedrick Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason A. Spector
- Laboratory of Bioregenerative Medicine and Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lewis C. Cantley
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
24
|
Senolytic effect of high intensity interval exercise on human skeletal muscle. Aging (Albany NY) 2023; 15:765-776. [PMID: 36779839 PMCID: PMC9970302 DOI: 10.18632/aging.204511] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023]
Abstract
p16INK4a expression is a robust biomarker of senescence for stem cells in human tissues. Here we examined the effect of exercise intensity on in vivo senescence in skeletal muscle, using a randomized counter-balanced crossover design. Biopsied vastus lateralis of 9 sedentary men (age 26.1 ± 2.5 y) were assessed before and after a single bout of moderate steady state exercise (SSE, 60% maximal aerobic power) and high intensity interval exercise (HIIE, 120% maximal aerobic power) on a cycloergometer accumulating same amount of cycling work (in kilojoule). Increases in cell infiltration (+1.2 folds), DNA strand break (+1.3 folds), and γ-H2AX+ myofibers (+1.1 folds) occurred immediately after HIIE and returned to baseline in 24 h (p < 0.05). Muscle p16Ink4a mRNA decreased 24 h after HIIE (-57%, p < 0.05). SSE had no effect on cell infiltration, p16Ink4a mRNA, and DNA strand break in muscle tissues. Senescence-lowering effect of HIIE was particularly prominent in the muscle with high pre-exercise p16INK4a expression, suggesting that exercise intensity determines the level of selection pressure to tissue stem cells at late senescent stage in human skeletal muscle. This evidence provides an explanation for the discrepancy between destructive nature of high intensity exercise and its anti-aging benefits.
Collapse
|
25
|
Santos AL, Sinha S. Ageing, Metabolic Dysfunction, and the Therapeutic Role of Antioxidants. Subcell Biochem 2023; 103:341-435. [PMID: 37120475 DOI: 10.1007/978-3-031-26576-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The gradual ageing of the world population has been accompanied by a dramatic increase in the prevalence of obesity and metabolic diseases, especially type 2 diabetes. The adipose tissue dysfunction associated with ageing and obesity shares many common physiological features, including increased oxidative stress and inflammation. Understanding the mechanisms responsible for adipose tissue dysfunction in obesity may help elucidate the processes that contribute to the metabolic disturbances that occur with ageing. This, in turn, may help identify therapeutic targets for the treatment of obesity and age-related metabolic disorders. Because oxidative stress plays a critical role in these pathological processes, antioxidant dietary interventions could be of therapeutic value for the prevention and/or treatment of age-related diseases and obesity and their complications. In this chapter, we review the molecular and cellular mechanisms by which obesity predisposes individuals to accelerated ageing. Additionally, we critically review the potential of antioxidant dietary interventions to counteract obesity and ageing.
Collapse
Affiliation(s)
- Ana L Santos
- IdISBA - Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain.
| | | |
Collapse
|
26
|
A Glb1-2A-mCherry reporter monitors systemic aging and predicts lifespan in middle-aged mice. Nat Commun 2022; 13:7028. [PMID: 36396643 PMCID: PMC9671911 DOI: 10.1038/s41467-022-34801-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
The progressive decline of physiological function and the increased risk of age-related diseases challenge healthy aging. Multiple anti-aging manipulations, such as senolytics, have proven beneficial for health; however, the biomarkers that label in vivo senescence at systemic levels are lacking, thus hindering anti-aging applications. In this study, we generate a Glb1+/m‒Glb1-2A-mCherry (GAC) reporter allele at the Glb1 gene locus, which encodes lysosomal β-galactosidase-an enzyme elevated in tissues of old mice. A linear correlation between GAC signal and chronological age is established in a cohort of middle-aged (9 to 13 months) Glb1+/m mice. The high GAC signal is closely associated with cardiac hypertrophy and a shortened lifespan. Moreover, the GAC signal is exponentially increased in pathological senescence induced by bleomycin in the lung. Senolytic dasatinib and quercetin (D + Q) reduce GAC signal in bleomycin treated mice. Thus, the Glb1-2A-mCherry reporter mice monitors systemic aging and function decline, predicts lifespan, and may facilitate the understanding of aging mechanisms and help in the development of anti-aging interventions.
Collapse
|
27
|
From cyclins to CDKIs: Cell cycle regulation of skeletal muscle stem cell quiescence and activation. Exp Cell Res 2022; 420:113275. [PMID: 35931143 DOI: 10.1016/j.yexcr.2022.113275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/12/2022] [Accepted: 07/03/2022] [Indexed: 11/22/2022]
Abstract
After extensive proliferation during development, the adult skeletal muscle cells remain outside the cell cycle, either as post-mitotic myofibers or as quiescent muscle stem cells (MuSCs). Despite its terminally differentiated state, adult skeletal muscle has a remarkable regeneration potential, driven by MuSCs. Upon injury, MuSC quiescence is reversed to support tissue growth and repair and it is re-established after the completion of muscle regeneration. The distinct cell cycle states and transitions observed in the different myogenic populations are orchestrated by elements of the cell cycle machinery. This consists of i) complexes of cyclins and Cyclin-Dependent Kinases (CDKs) that ensure cell cycle progression and ii) their negative regulators, the Cyclin-Dependent Kinase Inhibitors (CDKIs). In this review we discuss the roles of these factors in developmental and adult myogenesis, with a focus on CDKIs that have emerging roles in stem cell functions.
Collapse
|
28
|
Dungan CM, Figueiredo VC, Wen Y, VonLehmden GL, Zdunek CJ, Thomas NT, Mobley CB, Murach KA, Brightwell CR, Long DE, Fry CS, Kern PA, McCarthy JJ, Peterson CA. Senolytic treatment rescues blunted muscle hypertrophy in old mice. GeroScience 2022; 44:1925-1940. [PMID: 35325353 PMCID: PMC9616988 DOI: 10.1007/s11357-022-00542-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 03/06/2022] [Indexed: 01/07/2023] Open
Abstract
With aging, skeletal muscle plasticity is attenuated in response to exercise. Here, we report that senescent cells, identified using senescence-associated β-galactosidase (SA β-Gal) activity and p21 immunohistochemistry, are very infrequent in resting muscle, but emerge approximately 2 weeks after a bout of resistance exercise in humans. We hypothesized that these cells contribute to blunted hypertrophic potential in old age. Using synergist ablation-induced mechanical overload (MOV) of the plantaris muscle to model resistance training in adult (5-6-month) and old (23-24-month) male C57BL/6 J mice, we found increased senescent cells in both age groups during hypertrophy. Consistent with the human data, there were negligible senescent cells in plantaris muscle from adult and old sham controls, but old mice had significantly more senescent cells 7 and 14 days following MOV relative to young. Old mice had blunted whole-muscle hypertrophy when compared to adult mice, along with smaller muscle fibers, specifically glycolytic type 2x + 2b fibers. To ablate senescent cells using a hit-and-run approach, old mice were treated with vehicle or a senolytic cocktail consisting of 5 mg/kg dasatinib and 50 mg/kg quercetin (D + Q) on days 7 and 10 during 14 days of MOV; control mice underwent sham surgery with or without senolytic treatment. Old mice given D + Q had larger muscles and muscle fibers after 14 days of MOV, fewer senescent cells when compared to vehicle-treated old mice, and changes in the expression of genes (i.e., Igf1, Ddit4, Mmp14) that are associated with hypertrophic growth. Our data collectively show that senescent cells emerge in human and mouse skeletal muscle following a hypertrophic stimulus and that D + Q improves muscle growth in old mice.
Collapse
Affiliation(s)
- Cory M Dungan
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA.
- College of Health Sciences, University of Kentucky, 900 S. Limestone, CTW 445, Lexington, KY, 40536, USA.
| | | | - Yuan Wen
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | | | | | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - C Brooks Mobley
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- School of Kinesiology, Auburn University, Auburn, AL, USA
| | - Kevin A Murach
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Camille R Brightwell
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Douglas E Long
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Philip A Kern
- Department of Internal Medicine, Division of Endocrinology, University of Kentucky, Lexington, KY, USA
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Charlotte A Peterson
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
29
|
Zhang X, Habiballa L, Aversa Z, Ng YE, Sakamoto AE, Englund DA, Pearsall VM, White TA, Robinson MM, Rivas DA, Dasari S, Hruby AJ, Lagnado AB, Jachim SK, Granic A, Sayer AA, Jurk D, Lanza IR, Khosla S, Fielding RA, Nair KS, Schafer MJ, Passos JF, LeBrasseur NK. Characterization of cellular senescence in aging skeletal muscle. NATURE AGING 2022; 2:601-615. [PMID: 36147777 PMCID: PMC9491365 DOI: 10.1038/s43587-022-00250-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/08/2022] [Indexed: 01/10/2023]
Abstract
Senescence is a cell fate that contributes to multiple aging-related pathologies. Despite profound age-associated changes in skeletal muscle (SkM), whether its constituent cells are prone to senesce has not been methodically examined. Herein, using single cell and bulk RNA-sequencing and complementary imaging methods on SkM of young and old mice, we demonstrate that a subpopulation of old fibroadipogenic progenitors highly expresses p16 Ink4a together with multiple senescence-related genes and, concomitantly, exhibits DNA damage and chromatin reorganization. Through analysis of isolated myofibers, we also detail a senescence phenotype within a subset of old cells, governed instead by p2 Cip1 . Administration of a senotherapeutic intervention to old mice countered age-related molecular and morphological changes and improved SkM strength. Finally, we found that the senescence phenotype is conserved in SkM from older humans. Collectively, our data provide compelling evidence for cellular senescence as a hallmark and potentially tractable mediator of SkM aging.
Collapse
Affiliation(s)
- Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
- These authors equally contributed to this work
| | - Leena Habiballa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Mayo clinic graduate school of biomedical science, rochester, MN, USA
- These authors equally contributed to this work
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Yan Er Ng
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Ayumi E. Sakamoto
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Davis A. Englund
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Thomas A. White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Matthew M. Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Donato A. Rivas
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center, Tufts University, Boston, Massachusetts, USA
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Adam J. Hruby
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Anthony B. Lagnado
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Sarah K. Jachim
- Mayo clinic graduate school of biomedical science, rochester, MN, USA
| | - Antoneta Granic
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Avan A. Sayer
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ian R. Lanza
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Roger A. Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center, Tufts University, Boston, Massachusetts, USA
| | - K. Sreekumaran Nair
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Marissa J. Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Taivassalo T, Hepple RT. Integrating Mechanisms of Exacerbated Atrophy and Other Adverse Skeletal Muscle Impact in COPD. Front Physiol 2022; 13:861617. [PMID: 35721564 PMCID: PMC9203961 DOI: 10.3389/fphys.2022.861617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
The normal decline in skeletal muscle mass that occurs with aging is exacerbated in patients with chronic obstructive pulmonary disease (COPD) and contributes to poor health outcomes, including a greater risk of death. There has been controversy about the causes of this exacerbated muscle atrophy, with considerable debate about the degree to which it reflects the very sedentary nature of COPD patients vs. being precipitated by various aspects of the COPD pathophysiology and its most frequent proximate cause, long-term smoking. Consistent with the latter view, recent evidence suggests that exacerbated aging muscle loss with COPD is likely initiated by decades of smoking-induced stress on the neuromuscular junction that predisposes patients to premature failure of muscle reinnervation capacity, accompanied by various alterations in mitochondrial function. Superimposed upon this are various aspects of COPD pathophysiology, such as hypercapnia, hypoxia, and inflammation, that can also contribute to muscle atrophy. This review will summarize the available knowledge concerning the mechanisms contributing to exacerbated aging muscle affect in COPD, consider the potential role of comorbidities using the specific example of chronic kidney disease, and identify emerging molecular mechanisms of muscle impairment, including mitochondrial permeability transition as a mechanism of muscle atrophy, and chronic activation of the aryl hydrocarbon receptor in driving COPD muscle pathophysiology.
Collapse
Affiliation(s)
- Tanja Taivassalo
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Russell T. Hepple
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States
- *Correspondence: Russell T. Hepple,
| |
Collapse
|
31
|
Maciel JIHN, Zazula MF, Rodrigues DFS, De Toni Boaro C, Boaretto ML, de Andrade BZ, Schneider SCS, Naliwaiko K, Torrejais MM, Costa RM, de Fátima Chasko Ribeiro L, Bertolini GRF. Whole-Body Vibration Promotes Skeletal Muscle Restructuring and Reduced Obesogenic Effect of MSG in Wistar Rats. Appl Biochem Biotechnol 2022; 194:3594-3608. [PMID: 35460454 DOI: 10.1007/s12010-022-03923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 11/02/2022]
Abstract
The negative changes of obesity to the locomotor system are a major concern in the current scenario, where obesity and metabolic syndrome are recurrent in Western societies. A physical exercise is an important tool as a way to rehabilitate obesity, highlighting whole-body vibration, as it is an easy-access modality with few restrictions. In this sense, we sought to evaluate the effect of whole-body vibration on the extensor digitorum longus muscle on a monosodium glutamate-induced obesity model. The main findings of the present study are related to the ability of the treatment with vibration to reduce the obesogenic characteristics and slow down the dyslipidemic condition of the animals. Likewise, the vibration promoted by the vibrating platform was essential in the recovery of the muscle structure, as well as the recovery of the muscle's oxidative capacity, initially compromised by obesity.
Collapse
Affiliation(s)
- Jhyslayne Ignácia Hoff Nunes Maciel
- Laboratório de Estudo das Lesões e Recursos Fisioterapêuticos, Universidade Estadual do Oeste do Paraná (Unioeste), Universitária St, 2069, Zip code: 85819110 , Paraná, Cascavel, Brasil
| | - Matheus Felipe Zazula
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Universidade Federal do Paraná, Curitiba, Paraná, Brasil
| | | | | | - Mariana Laís Boaretto
- Laboratório de Estudo das Lesões e Recursos Fisioterapêuticos, Universidade Estadual do Oeste do Paraná (Unioeste), Universitária St, 2069, Zip code: 85819110 , Paraná, Cascavel, Brasil
| | | | - Sara Cristina Sagae Schneider
- Laboratório de Estudo das Lesões e Recursos Fisioterapêuticos, Universidade Estadual do Oeste do Paraná (Unioeste), Universitária St, 2069, Zip code: 85819110 , Paraná, Cascavel, Brasil
| | - Katya Naliwaiko
- Laboratório de Plasticidade Morfofuncional, Departamento de Biologia Celular, Universidade Federal do Paraná, Curitiba, Paraná, Brasil
| | | | - Rose Meire Costa
- Laboratório de Biologia Estrutural e Funcional, Unioeste, Cascavel, Paraná, Brasil
| | | | - Gladson Ricardo Flor Bertolini
- Laboratório de Estudo das Lesões e Recursos Fisioterapêuticos, Universidade Estadual do Oeste do Paraná (Unioeste), Universitária St, 2069, Zip code: 85819110 , Paraná, Cascavel, Brasil.
| |
Collapse
|
32
|
Masteika IF, Sathya A, Homma S, Miller BM, Boyce FM, Miller JB. Downstream events initiated by expression of FSHD-associated DUX4: Studies of nucleocytoplasmic transport, γH2AX accumulation, and Bax/Bak-dependence. Biol Open 2022; 11:274475. [PMID: 35191484 PMCID: PMC8890089 DOI: 10.1242/bio.059145] [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: 11/15/2021] [Accepted: 01/12/2022] [Indexed: 12/15/2022] Open
Abstract
Abnormal expression in skeletal muscle of the double homeobox transcription factor DUX4 underlies pathogenesis in facioscapulohumeral muscular dystrophy (FSHD). Though multiple changes are known to be initiated by aberrant DUX4 expression, the downstream events initiated by DUX4 remain incompletely understood. In this study, we examined plausible downstream events initiated by DUX4. First, we found that nucleocytoplasmic protein export appeared to be decreased upon DUX4 expression as indicated by nuclear accumulation of a shuttle-GFP reporter. Second, building on studies from other labs, we showed that phospho(Ser139)-H2AX (γH2AX), an indicator of double-strand DNA breaks, accumulated both in human FSHD1 myotube nuclei upon endogenous DUX4 expression and in Bax-/-;Bak-/- (double knockout), SV40-immortalized mouse embryonic fibroblasts upon exogenous DUX4 expression. In contrast, DUX4-induced caspase 3/7 activation was prevented in Bax-/-;Bak-/- double knockout SV40-MEFs, but not by single knockouts of Bax, Bak, or Bid. Thus, aberrant DUX4 expression appeared to alter nucleocytoplasmic protein transport and generate double-strand DNA breaks in FSHD1 myotube nuclei, and the Bax/Bak pathway is required for DUX4-induced caspase activation but not γH2AX accumulation. These results add to our knowledge of downstream events induced by aberrant DUX4 expression and suggest possibilities for further mechanistic investigation.
Collapse
Affiliation(s)
- Isabel F Masteika
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Anvitha Sathya
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Sachiko Homma
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Bess M Miller
- Biological & Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Frederick M Boyce
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Jeffrey Boone Miller
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| |
Collapse
|
33
|
Dungan CM, Murach KA, Zdunek CJ, Tang ZJ, Nolt GL, Brightwell CR, Hettinger Z, Englund D, Liu Z, Fry CS, Filareto A, Franti M, Peterson CA. Deletion of SA β-Gal+ cells using senolytics improves muscle regeneration in old mice. Aging Cell 2022; 21:e13528. [PMID: 34904366 PMCID: PMC8761017 DOI: 10.1111/acel.13528] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/05/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022] Open
Abstract
Systemic deletion of senescent cells leads to robust improvements in cognitive, cardiovascular, and whole-body metabolism, but their role in tissue reparative processes is incompletely understood. We hypothesized that senolytic drugs would enhance regeneration in aged skeletal muscle. Young (3 months) and old (20 months) male C57Bl/6J mice were administered the senolytics dasatinib (5 mg/kg) and quercetin (50 mg/kg) or vehicle bi-weekly for 4 months. Tibialis anterior (TA) was then injected with 1.2% BaCl2 or PBS 7- or 28 days prior to euthanization. Senescence-associated β-Galactosidase positive (SA β-Gal+) cell abundance was low in muscle from both young and old mice and increased similarly 7 days following injury in both age groups, with no effect of D+Q. Most SA β-Gal+ cells were also CD11b+ in young and old mice 7- and 14 days following injury, suggesting they are infiltrating immune cells. By 14 days, SA β-Gal+/CD11b+ cells from old mice expressed senescence genes, whereas those from young mice expressed higher levels of genes characteristic of anti-inflammatory macrophages. SA β-Gal+ cells remained elevated in old compared to young mice 28 days following injury, which were reduced by D+Q only in the old mice. In D+Q-treated old mice, muscle regenerated following injury to a greater extent compared to vehicle-treated old mice, having larger fiber cross-sectional area after 28 days. Conversely, D+Q blunted regeneration in young mice. In vitro experiments suggested D+Q directly improve myogenic progenitor cell proliferation. Enhanced physical function and improved muscle regeneration demonstrate that senolytics have beneficial effects only in old mice.
Collapse
Affiliation(s)
- Cory M. Dungan
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Kevin A. Murach
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Present address:
Department of Health, Human Performance, and Recreation, and Cell and Molecular Biology ProgramUniversity of ArkansasFayettevilleArkansasUSA
| | | | - Zuo Jian Tang
- Computational BiologyGCBDSBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Georgia L. Nolt
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Camille R. Brightwell
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Athletic Training and Clinical NutritionCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Zachary Hettinger
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Davis A. Englund
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Zheng Liu
- Computational BiologyGCBDSBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Christopher S. Fry
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Athletic Training and Clinical NutritionCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Antonio Filareto
- Regenerative MedicineBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Michael Franti
- Regenerative MedicineBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Charlotte A. Peterson
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
34
|
Esteves de Lima J, Relaix F. Epigenetic Regulation of Myogenesis: Focus on the Histone Variants. Int J Mol Sci 2021; 22:ijms222312727. [PMID: 34884532 PMCID: PMC8657657 DOI: 10.3390/ijms222312727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 01/04/2023] Open
Abstract
Skeletal muscle development and regeneration rely on the successive activation of specific transcription factors that engage cellular fate, promote commitment, and drive differentiation. Emerging evidence demonstrates that epigenetic regulation of gene expression is crucial for the maintenance of the cell differentiation status upon division and, therefore, to preserve a specific cellular identity. This depends in part on the regulation of chromatin structure and its level of condensation. Chromatin architecture undergoes remodeling through changes in nucleosome composition, such as alterations in histone post-translational modifications or exchange in the type of histone variants. The mechanisms that link histone post-translational modifications and transcriptional regulation have been extensively evaluated in the context of cell fate and differentiation, whereas histone variants have attracted less attention in the field. In this review, we discuss the studies that have provided insights into the role of histone variants in the regulation of myogenic gene expression, myoblast differentiation, and maintenance of muscle cell identity.
Collapse
|
35
|
Saito Y, Chikenji TS. Diverse Roles of Cellular Senescence in Skeletal Muscle Inflammation, Regeneration, and Therapeutics. Front Pharmacol 2021; 12:739510. [PMID: 34552495 PMCID: PMC8450382 DOI: 10.3389/fphar.2021.739510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle undergoes vigorous tissue remodeling after injury. However, aging, chronic inflammatory diseases, sarcopenia, and neuromuscular disorders cause muscle loss and degeneration, resulting in muscular dysfunction. Cellular senescence, a state of irreversible cell cycle arrest, acts during normal embryonic development and remodeling after tissue damage; when these processes are complete, the senescent cells are eliminated. However, the accumulation of senescent cells is a hallmark of aging tissues or pathological contexts and may lead to progressive tissue degeneration. The mechanisms responsible for the effects of senescent cells have not been fully elucidated. Here, we review current knowledge about the beneficial and detrimental effects of senescent cells in tissue repair, regeneration, aging, and age-related disease, especially in skeletal muscle. We also discuss how senescence of muscle stem cells and muscle-resident fibro-adipogenic progenitors affects muscle pathologies or regeneration, and consider the possibility that immunosenescence leads to muscle pathogenesis. Finally, we explore senotherapy, the therapeutic targeting of senescence to treat age-related disease, from the standpoint of improving muscle regeneration.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
36
|
Lee TXY, Wu J, Jean WH, Condello G, Alkhatib A, Hsieh CC, Hsieh YW, Huang CY, Kuo CH. Reduced stem cell aging in exercised human skeletal muscle is enhanced by ginsenoside Rg1. Aging (Albany NY) 2021; 13:16567-16576. [PMID: 34181580 PMCID: PMC8266347 DOI: 10.18632/aging.203176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Background: Stem cell aging, characterized by elevated p16INK4a expression, decreases cell repopulating and self-renewal abilities, which results in elevated inflammation and slow recovery against stress. Methods: Biopsied muscles were analyzed at baseline and 24 h after squat exercise in 12 trained men (22 ± 2 y). Placebo (PLA) and immunostimulant Rg1 (5 mg) were supplemented 1 h before a squat exercise, using a double-blind counterbalanced crossover design. Results: Perceived exertion at the end of resistance exercise session was significantly lowered after Rg1 supplementation. Exercise doubled endothelial progenitor cells (EPC) (p < 0.001) and decreased p16INK4a mRNA to 50% of baseline (d = 0.865, p < 0.05) in muscle tissues, despite p16INK4a+ cell and beta-galactosidase+ (ß-Gal+) cell counts being unaltered. Rg1 further lowered p16INK4a mRNA to 35% of baseline with greater effect size than the PLA level (d = 1.302, p < 0.01) and decreased myeloperoxidase (MPO) mRNA to 39% of baseline (p < 0.05). A strong correlation between MPO and p16INK4a expression in muscle tissues was observed (r = 0.84, p < 0.001). Conclusion: EPC in skeletal muscle doubled 1 d after an acute bout of resistance exercise. The exercised effects in lowering EPC aging and tissue inflammation were enhanced by immunostimulant Rg1, suggesting the involvement of immune stimulation on EPC rejuvenation.
Collapse
Affiliation(s)
- Tania Xu Yar Lee
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC
| | - Jinfu Wu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC.,Laboratory of Regenerative Medicine in Sports Science, School of Physical Education & Sports Science, South China Normal University, Guangzhou, China
| | - Wei-Horng Jean
- Department of Anesthesiology, Far East Memorial Hospital, New Taipei City 220, Taiwan, ROC
| | - Giancarlo Condello
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC
| | - Ahmad Alkhatib
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC.,School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, England, United Kingdom
| | - Chao-Chieh Hsieh
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC
| | - Yu-Wen Hsieh
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan, ROC.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, ROC.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan, ROC
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei City 11153, Taiwan, ROC
| |
Collapse
|
37
|
Bhardwaj P, Brown KA. Obese Adipose Tissue as a Driver of Breast Cancer Growth and Development: Update and Emerging Evidence. Front Oncol 2021; 11:638918. [PMID: 33859943 PMCID: PMC8042134 DOI: 10.3389/fonc.2021.638918] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an established risk factor for breast cancer growth and progression. A number of advances have been made in recent years revealing new insights into this link. Early events in breast cancer development involve the neoplastic transformation of breast epithelial cells to cancer cells. In obesity, breast adipose tissue undergoes significant hormonal and inflammatory changes that create a mitogenic microenvironment. Many factors that are produced in obesity have also been shown to promote tumorigenesis. Given that breast epithelial cells are surrounded by adipose tissue, the crosstalk between the adipose compartment and breast epithelial cells is hypothesized to be a significant player in the initiation and progression of breast cancer in individuals with excess adiposity. The present review examines this crosstalk with a focus on obese breast adipose-derived estrogen, inflammatory mediators and adipokines, and how they are mechanistically linked to breast cancer risk and growth through stimulation of oxidative stress, DNA damage, and pro-oncogenic transcriptional programs. Pharmacological and lifestyle strategies targeting these factors and their downstream effects are evaluated for feasibility and efficacy in decreasing the risk of obesity-induced breast epithelial cell transformation and consequently, breast cancer development.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
38
|
Regeneration during Obesity: An Impaired Homeostasis. Animals (Basel) 2020; 10:ani10122344. [PMID: 33317011 PMCID: PMC7763812 DOI: 10.3390/ani10122344] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Regeneration represents the biological processes that allow cells and tissues to renew and develop. During obesity, a variety of changes and reactions are seen. This includes inflammation and metabolic disorders. These obesity-induced changes do impact the regeneration processes. Such impacts that obesity has on regeneration would affect tissues and organs development and would also have consequences on the outcomes of therapies that depend on cells regeneration (such as burns, radiotherapy and leukemia) given to patients suffering from obesity. Therefore, a particular attention should be given to patients suffering from obesity in biological, therapeutic and clinical contexts that depend on regeneration ability. Abstract Obesity is a health problem that, in addition to the known morbidities, induces the generation of a biological environment with negative impacts on regeneration. Indeed, factors like DNA damages, oxidative stress and inflammation would impair the stem cell functions, in addition to some metabolic and development patterns. At the cellular and tissulaire levels, this has consequences on growth, renewal and restoration which results into an impaired regeneration. This impaired homeostasis concerns also key metabolic tissues including muscles and liver which would worsen the energy balance outcome towards further development of obesity. Such impacts of obesity on regeneration shows the need of a specific care given to obese patients recovering from diseases or conditions requiring regeneration such as burns, radiotherapy and leukemia. On the other hand, since stem cells are suggested to manage obesity, this impaired regeneration homeostasis needs to be considered towards more optimized stem cells-based obesity therapies within the context of precision medicine.
Collapse
|
39
|
Kletzien H, Kelm-Nelson CA, Wang S, Suzuki M, Connor NP. Myogenic marker expression as a function of age and exercise-based therapy in the tongue. Exp Gerontol 2020; 142:111104. [PMID: 33017670 PMCID: PMC7748063 DOI: 10.1016/j.exger.2020.111104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/29/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022]
Abstract
Degeneration of tongue muscles with aging may contribute to swallowing deficits observed in elderly people. However, the capacity for tongue muscle stem cells (SCs) to regenerate and repair the aged tongue and improve tongue strength following tongue exercise (a current clinical treatment) has never been examined. We found that the expression of regenerative, myogenic markers were impaired with age and may be related to increased expression of senescent marker p16INK4a. Tongue strength increased in young adult and old rats following exercise and was related to the expression of Pax7, MyoD, myogenin, and p16INK4a. Our study also suggests that strengthening of tongue muscles via clinical rehabilitation strategies also increased the expression of SC regenerative markers in the tongue throughout the exercise duration.
Collapse
Affiliation(s)
- Heidi Kletzien
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States of America; Department of Surgery, University of Wisconsin School of Medicine and Public Health, United States of America; Department of Stem Cell and Regenerative Biology, Harvard University, United States of America.
| | - Cynthia A Kelm-Nelson
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, United States of America
| | - Sabrina Wang
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, United States of America
| | - Masatoshi Suzuki
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States of America; Department of Comparative Biosciences, University of Wisconsin-Madison, United States of America
| | - Nadine P Connor
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, United States of America; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, United States of America
| |
Collapse
|
40
|
Aerobic exercise induces tumor suppressor p16 INK4a expression of endothelial progenitor cells in human skeletal muscle. Aging (Albany NY) 2020; 12:20226-20234. [PMID: 33104519 PMCID: PMC7655215 DOI: 10.18632/aging.103763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/07/2020] [Indexed: 12/28/2022]
Abstract
Aerobic exercise induces oxidative stress and DNA damage, nevertheless, lowers cancer incidence. It remains unclear how genetic stability is maintained under this condition. Here, we examined the dynamic change of the tumor suppressor p16INK4a in cells of skeletal muscle among young men following 60-min of aerobic cycling at 70% maximal oxygen consumption (V̇O2max). Rg1 (5 mg, an immunostimulant ginsenoside) and placebo (PLA) were supplemented 1 h before exercise. Data from serial muscle biopsies shows unchanged p16INK4a+ cells after exercise followed by a considerable increase (+21-fold) in vastus lateralis muscle 3 h later. This increase was due to the accumulation of endothelial progenitor cells (p16INK4a+/CD34+) surrounding myofibers and other infiltrated nucleated cells (p16INK4a+/CD34-) in necrotic myofibers. During the Rg1 trial, acute increases of p16INK4a+ cells in the muscle occurred immediately after exercise (+3-fold) and reversed near baseline 3 h later. Rg1 also lowered IL-10 mRNA relative to PLA 3 h after exercise. Post-exercise increases in VEGF mRNA and CD163+ macrophages were similar for PLA and Rg1 trials. Conclusion: The marked increases in p16INK4a protein expression of endothelial progenitor cells in skeletal muscle implicates a protective mechanism for maintaining genetic stability against aerobic exercise. Rg1 accelerates resolution of the exercise-induced stress response.
Collapse
|
41
|
Regulation of microRNAs in Satellite Cell Renewal, Muscle Function, Sarcopenia and the Role of Exercise. Int J Mol Sci 2020; 21:ijms21186732. [PMID: 32937893 PMCID: PMC7555198 DOI: 10.3390/ijms21186732] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Sarcopenia refers to a condition of progressive loss of skeletal muscle mass and function associated with a higher risk of falls and fractures in older adults. Musculoskeletal aging leads to reduced muscle mass and strength, affecting the quality of life in elderly people. In recent years, several studies contributed to improve the knowledge of the pathophysiological alterations that lead to skeletal muscle dysfunction; however, the molecular mechanisms underlying sarcopenia are still not fully understood. Muscle development and homeostasis require a fine gene expression modulation by mechanisms in which microRNAs (miRNAs) play a crucial role. miRNAs modulate key steps of skeletal myogenesis including satellite cells renewal, skeletal muscle plasticity, and regeneration. Here, we provide an overview of the general aspects of muscle regeneration and miRNAs role in skeletal mass homeostasis and plasticity with a special interest in their expression in sarcopenia and skeletal muscle adaptation to exercise in the elderly.
Collapse
|