1
|
Karami E, Bazgir B, Shirvani H, Mohammadi MT, Khaledi M. Unraveling the bidirectional relationship between muscle inflammation and satellite cells activity: influencing factors and insights. J Muscle Res Cell Motil 2024:10.1007/s10974-024-09683-7. [PMID: 39508952 DOI: 10.1007/s10974-024-09683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Inflammation stands as a vital and innate function of the immune system, essential for maintaining physiological homeostasis. Its role in skeletal muscle regeneration is pivotal, with the activation of satellite cells (SCs) driving the repair and generation of new myofibers. However, the relationship between inflammation and SCs is intricate, influenced by various factors. Muscle injury and repair prompt significant infiltration of immune cells, particularly macrophages, into the muscle tissue. The interplay of cytokines and chemokines from diverse cell types, including immune cells, fibroadipogenic progenitors, and SCs, further shapes the inflammation-SCs dynamic. While some studies suggest heightened inflammation associates with reduced SC activity and increased fibro- or adipogenesis, others indicate an inflammatory stimulus benefits SC function. Yet, the existing literature struggles to delineate clearly between the stimulatory and inhibitory effects of inflammation on SCs and muscle regeneration. This paper comprehensively reviews studies exploring the impact of pharmacological agents, dietary interventions, genetic factors, and exercise regimes on the interplay between inflammation and SC activity.
Collapse
Affiliation(s)
- Esmail Karami
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Bazgir
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mansoor Khaledi
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
2
|
Lee EJ, Charles JF, Sinha I, Neppl RL. Loss of HNRNPU in Skeletal Muscle Increases Intramuscular Infiltration of Ly6C Positive Cells, leading to Muscle Atrophy through Activation of NF-κB Signaling. Adv Biol (Weinh) 2024; 8:e2400152. [PMID: 38797891 DOI: 10.1002/adbi.202400152] [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: 03/18/2024] [Revised: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Heterogeneous nuclear ribonucleoprotein U (hnRNPU) is known to play multiple biological roles by regulating transcriptional expression, RNA splicing, RNA stability, and chromatin structure in a tissue-dependent manner. The role of hnRNPU in skeletal muscle development and maintenance has not been previously evaluated. In this study, skeletal muscle specific hnRNPU knock out mice is utilized and evaluated skeletal muscle mass and immune cell infiltration through development. By 4 weeks, muscle-specific hnRNPU knockout mice revealed Ly6C+ monocyte infiltration into skeletal muscle, which preceded muscle atrophy. Canonical NF-kB signaling is activated in a myofiber-autonomous manner with hnRNPU repression. Inducible hnRNPU skeletal muscle knockout mice further demonstrated that deletion of hnRNPU in adulthood is sufficient to cause muscle atrophy, suggesting that hnRNPU's role in muscle maintenance is not during development alone. Treatment with salirasib, to inhibit proliferation of immune cells, prevents muscle atrophy in muscle-specific hnRNPU knock out mice, indicating that immune cell infiltration plays causal role in muscle atrophy of hnRNPU knock out mice. Overall, the findings suggest that loss of hnRNPU triggers muscle inflammation and activates NF-κB signaling in a cell-autonomous manner, culminating in muscle atrophy.
Collapse
Affiliation(s)
- Eun-Joo Lee
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Julia F Charles
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Indranil Sinha
- Division of Plastic and reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ronald L Neppl
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Fioretti D, Ledda M, Iurescia S, Carletti R, Di Gioia C, Lolli MG, Marchese R, Lisi A, Rinaldi M. Severely Damaged Freeze-Injured Skeletal Muscle Reveals Functional Impairment, Inadequate Repair, and Opportunity for Human Stem Cell Application. Biomedicines 2023; 12:30. [PMID: 38275391 PMCID: PMC10813063 DOI: 10.3390/biomedicines12010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. MATERIALS AND METHODS We caused a freeze injury (FI) in the biceps femoris of C57BL/6N mice. From day 1 to day 25 post-injury, we conducted histological/morphometric examinations, an analysis of the expression of genes involved in inflammation/regeneration, and an in vivo functional evaluation. RESULTS We found that FI activates cytosolic DNA sensing and inflammatory responses. Persistent macrophage infiltration, the prolonged expression of eMHC, the presence of centrally nucleated myofibers, and the presence of PAX7+ satellite cells at late time points and with chronic physical impairment indicated inadequate repair. By looking at stem-cell-based therapeutic protocols of muscle repair, we investigated the crosstalk between M1-biased macrophages and human amniotic mesenchymal stem cells (hAMSCs) in vitro. We demonstrated their reciprocal paracrine effects where hAMSCs induced a shift of M1 macrophages into an anti-inflammatory phenotype, and M1 macrophages promoted an increase in the expression of hAMSC immunomodulatory factors. CONCLUSIONS Our findings support the rationale for the future use of our injury model to exploit the full potential of in vivo hAMSC transplantation following severe traumatic injuries.
Collapse
Affiliation(s)
- Daniela Fioretti
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Mario Ledda
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Sandra Iurescia
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Cira Di Gioia
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Grazia Lolli
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Rodolfo Marchese
- Department of Clinical Pathology, FBF S. Peter Hospital, 00189 Rome, Italy;
| | - Antonella Lisi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Monica Rinaldi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| |
Collapse
|
4
|
Wang Y, Sun Y, Yang C, Han B, Wang S. Sodium salicylate ameliorates exercise-induced muscle damage in mice by inhibiting NF-kB signaling. J Orthop Surg Res 2023; 18:967. [PMID: 38098039 PMCID: PMC10722820 DOI: 10.1186/s13018-023-04433-w] [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: 07/28/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Eccentric muscle contraction can cause muscle damage, which reduces the efficiency of exercise. Previous evidence suggested that Sodium salicylate (SS) could improve the repair of aged muscle. This study intends to investigate whether SS can impact skeletal muscle damage caused by eccentric exercise. METHODS Eccentric treadmill exercise was performed to induce muscle damage in mice. Plasma levels of muscle damage markers were estimated. RT-qPCR was employed for detecting mRNA levels of proinflammatory mediators in murine gastrocnemius muscle. Immunofluorescence staining of laminin/DAPI was utilized for quantifying centrally nucleated myofibers in the gastrocnemius muscle. Western blotting was implemented to examine protein levels of mitsugumin 53 (MG53), matrix metalloproteinase (MMP)-2/9, and NF-κB signaling-related markers. RESULTS SS administration reduced muscle damage marker production in the plasma and decreased the levels of proinflammatory mediators, MG53 and MMP-2/9 in mice after exercise. SS alleviated the severity of muscle damage in the gastrocnemius of mice after eccentric exercise. SS blocked NF-κB signaling pathway in the gastrocnemius muscle. CONCLUSION SS administration ameliorates skeletal muscle damage caused by eccentric exercise in the mouse model.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Sports, Northeastern University, Lane 3, Wenhua Road, Heping District, Shenyang City, 110819, China.
| | - Yuning Sun
- Department of Sports, Northeastern University, Lane 3, Wenhua Road, Heping District, Shenyang City, 110819, China
| | - Chunhui Yang
- Department of Sports, Northeastern University, Lane 3, Wenhua Road, Heping District, Shenyang City, 110819, China
| | - Bing Han
- Department of Sports, Northeastern University, Lane 3, Wenhua Road, Heping District, Shenyang City, 110819, China
| | - Sining Wang
- Department of General, Huanggu District People's Government Office, Shenyang City, 110032, China
| |
Collapse
|
5
|
Endo Y, Hwang CD, Zhang Y, Olumi S, Koh DJ, Zhu C, Neppl RL, Agarwal S, Sinha I. VEGFA Promotes Skeletal Muscle Regeneration in Aging. Adv Biol (Weinh) 2023; 7:e2200320. [PMID: 36988414 PMCID: PMC10539483 DOI: 10.1002/adbi.202200320] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/06/2023] [Indexed: 03/30/2023]
Abstract
Aging is associated with loss of skeletal muscle regeneration. Differentially regulated vascular endothelial growth factor (VEGF)A with aging may partially underlies this loss of regenerative capacity. To assess the role of VEGFA in muscle regeneration, young (12-14 weeks old) and old C57BL/6 mice (24,25 months old) are subjected to cryoinjury in the tibialis anterior (TA) muscle to induce muscle regeneration. The average cross-sectional area (CSA) of regenerating myofibers is 33% smaller in old as compared to young (p < 0.01) mice, which correlates with a two-fold loss of muscle VEGFA protein levels (p = 0.02). The capillary density in the TA is similar between the two groups. Young VEGFlo mice, with a 50% decrease in systemic VEGFA activity, exhibit a two-fold reduction in the average regenerating fiber CSA following cryoinjury (p < 0.01) in comparison to littermate controls. ML228, a hypoxia signaling activator known to increase VEGFA levels, augments muscle VEGFA levels and increases average CSA of regenerating fibers in both old mice (25% increase, p < 0.01) and VEGFlo (20% increase, p < 0.01) mice, but not in young or littermate controls. These results suggest that VEGFA may be a therapeutic target in age-related muscle loss.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Charles D. Hwang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Yuteng Zhang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Shayan Olumi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Daniel J. Koh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Christina Zhu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Ronald L. Neppl
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02114
| | - Shailesh Agarwal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| | - Indranil Sinha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard University, Boston, MA, 02115
| |
Collapse
|
6
|
Liu HW, Chang SJ. Effects of green tea-derived natural products on resistance exercise training in sarcopenia: A retrospective narrative mini-review. J Food Drug Anal 2023; 31:381-386. [PMID: 39666283 PMCID: PMC10629911 DOI: 10.38212/2224-6614.3470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/30/2023] [Indexed: 12/13/2024] Open
Abstract
Skeletal muscle function deficits result in metabolic disease development and physical dysfunction in older adults. Sarcopenia is characterized by a decrease in muscle mass and strength with advancing age, and it increases the risks of mobility impairments, disease development, and mortality. Lifestyle interventions involving a combination of diet and exercise to prevent and attenuate sarcopenia warrant substantial research attention. Resistance exercise training under supervision is a safe and the most effective approach to reducing age-related muscle loss and improving multiple aspects of overall health in the older population. The beneficial effects of resistance exercise training on skeletal muscle mass may be augmented by specific dietary supplements (i.e., green tea-derived natural products). The purpose of this mini review is to provide an up-to-date, evidence-based account of the effectiveness of green tea-derived natural products for supporting resistance training-induced adaptations to prevent or attenuate age-related muscle mass loss. Based on animal and clinical studies, we provide insights into supplementation with green tea-derived natural products, which may assist in the growth or maintenance of skeletal muscle and subsequently delay the onset of age-related metabolic diseases in older adults.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Physical Education and Sport Sciences, National Taiwan Normal University, Taipei,
Taiwan
| | - Sue-Joan Chang
- Department of Life Sciences, National Cheng Kung University, Tainan,
Taiwan
| |
Collapse
|
7
|
Bodine SC, Sinha I, Sweeney HL. Mechanisms of Skeletal Muscle Atrophy and Molecular Circuitry of Stem Cell Fate in Skeletal Muscle Regeneration and Aging. J Gerontol A Biol Sci Med Sci 2023; 78:14-18. [PMID: 37325966 PMCID: PMC10272973 DOI: 10.1093/gerona/glad023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 06/17/2023] Open
Abstract
Skeletal muscle is a complex and highly adaptable tissue. With aging, there is a progressive loss of muscle mass and function, known as sarcopenia, and a reduced capacity for regeneration and repair following injury. A review of the literature shows that the primary mechanisms underlying the age-related loss of muscle mass and the attenuated growth response are multi-factorial and related to alterations in multiple processes, including proteostasis, mitochondrial function, extracellular matrix remodeling, and neuromuscular junction function. Multiple factors influence the rate of sarcopenia, including acute illness and trauma, followed by incomplete recovery and repair. Regeneration and repair of damaged skeletal muscle involve an orchestrated cross-talk between multiple cell populations, including satellite cells, immune cells, and fibro-adipogenic precursor cells. Proof-of-concept studies in mice have demonstrated that reprogramming of this disrupted orchestration, resulting in the normalization of muscle function, may be possible using small molecules that target muscle macrophages. During aging, as well as in muscular dystrophies, disruptions in multiple signaling pathways and in the cross-talk between different cell populations contribute to the failure to properly repair and maintain muscle mass and function.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, and Iowa City VA Health Care System, Iowa City, Iowa, USA
| | - Indranil Sinha
- Department of Surgery, Division of Plastic Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Hugh Lee Sweeney
- University of Florida Myology Institute and Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
8
|
Farahzadi R, Valipour B, Montazersaheb S, Fathi E. Targeting the stem cell niche micro-environment as therapeutic strategies in aging. Front Cell Dev Biol 2023; 11:1162136. [PMID: 37274742 PMCID: PMC10235764 DOI: 10.3389/fcell.2023.1162136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Adult stem cells (ASCs) reside throughout the body and support various tissue. Owing to their self-renewal capacity and differentiation potential, ASCs have the potential to be used in regenerative medicine. Their survival, quiescence, and activation are influenced by specific signals within their microenvironment or niche. In better words, the stem cell function is significantly influenced by various extrinsic signals derived from the niche. The stem cell niche is a complex and dynamic network surrounding stem cells that plays a crucial role in maintaining stemness. Studies on stem cell niche have suggested that aged niche contributes to the decline in stem cell function. Notably, functional loss of stem cells is highly associated with aging and age-related disorders. The stem cell niche is comprised of complex interactions between multiple cell types. Over the years, essential aspects of the stem cell niche have been revealed, including cell-cell contact, extracellular matrix interaction, soluble signaling factors, and biochemical and biophysical signals. Any alteration in the stem cell niche causes cell damage and affects the regenerative properties of the stem cells. A pristine stem cell niche might be essential for the proper functioning of stem cells and the maintenance of tissue homeostasis. In this regard, niche-targeted interventions may alleviate problems associated with aging in stem cell behavior. The purpose of this perspective is to discuss recent findings in the field of stem cell aging, heterogeneity of stem cell niches, and impact of age-related changes on stem cell behavior. We further focused on how the niche affects stem cells in homeostasis, aging, and the progression of malignant diseases. Finally, we detail the therapeutic strategies for tissue repair, with a particular emphasis on aging.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
9
|
Tavakoli S, Garcia V, Gähwiler E, Adatto I, Rangan A, Messemer KA, Kakhki SA, Yang S, Chan VS, Manning ME, Fotowat H, Zhou Y, Wagers AJ, Zon LI. Transplantation-based screen identifies inducers of muscle progenitor cell engraftment across vertebrate species. Cell Rep 2023; 42:112365. [PMID: 37018075 PMCID: PMC10548355 DOI: 10.1016/j.celrep.2023.112365] [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: 10/21/2022] [Revised: 01/06/2023] [Accepted: 03/22/2023] [Indexed: 04/06/2023] Open
Abstract
Stem cell transplantation presents a potentially curative strategy for genetic disorders of skeletal muscle, but this approach is limited by the deleterious effects of cell expansion in vitro and consequent poor engraftment efficiency. In an effort to overcome this limitation, we sought to identify molecular signals that enhance the myogenic activity of cultured muscle progenitors. Here, we report the development and application of a cross-species small-molecule screening platform employing zebrafish and mice, which enables rapid, direct evaluation of the effects of chemical compounds on the engraftment of transplanted muscle precursor cells. Using this system, we screened a library of bioactive lipids to discriminate those that could increase myogenic engraftment in vivo in zebrafish and mice. This effort identified two lipids, lysophosphatidic acid and niflumic acid, both linked to the activation of intracellular calcium-ion flux, which showed conserved, dose-dependent, and synergistic effects in promoting muscle engraftment across these vertebrate species.
Collapse
Affiliation(s)
- Sahar Tavakoli
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Vivian Garcia
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Eric Gähwiler
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Institute for Regenerative Medicine, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Isaac Adatto
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Apoorva Rangan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Stanford Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kathleen A Messemer
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sara Ashrafi Kakhki
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Song Yang
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Victoria S Chan
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Margot E Manning
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Haleh Fotowat
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Yi Zhou
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA; Joslin Diabetes Center, Boston, MA 02215, USA.
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Sousa NS, Brás MF, Antunes IB, Lindholm P, Neves J, Sousa-Victor P. Aging disrupts MANF-mediated immune modulation during skeletal muscle regeneration. NATURE AGING 2023; 3:585-599. [PMID: 37118549 DOI: 10.1038/s43587-023-00382-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/16/2023] [Indexed: 04/30/2023]
Abstract
Age-related decline in skeletal muscle regenerative capacity is multifactorial, yet the contribution of immune dysfunction to regenerative failure is unknown. Macrophages are essential for effective debris clearance and muscle stem cell activity during muscle regeneration, but the regulatory mechanisms governing macrophage function during muscle repair are largely unexplored. Here, we uncover a new mechanism of immune modulation operating during skeletal muscle regeneration that is disrupted in aged animals and relies on the regulation of macrophage function. The immune modulator mesencephalic astrocyte-derived neurotrophic factor (MANF) is induced following muscle injury in young mice but not in aged animals, and its expression is essential for regenerative success. Regenerative impairments in aged muscle are associated with defects in the repair-associated myeloid response similar to those found in MANF-deficient models and could be improved through MANF delivery. We propose that restoring MANF levels is a viable strategy to improve myeloid response and regenerative capacity in aged muscle.
Collapse
Affiliation(s)
- Neuza S Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Margarida F Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Inês B Antunes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joana Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Sousa-Victor
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
11
|
McNamara SL, Seo BR, Freedman BR, Roloson EB, Alvarez JT, O'Neill CT, Vandenburgh HH, Walsh CJ, Mooney DJ. Anti-inflammatory therapy enables robot-actuated regeneration of aged muscle. Sci Robot 2023; 8:eadd9369. [PMID: 36947599 DOI: 10.1126/scirobotics.add9369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Robot-actuated mechanical loading (ML)-based therapies ("mechanotherapies") can promote regeneration after severe skeletal muscle injury, but the effectiveness of such approaches during aging is unknown and may be influenced by age-associated decline in the healing capacity of skeletal muscle. To address this knowledge gap, this work used a noninvasive, load-controlled robotic device to impose highly defined tissue stresses to evaluate the age dependence of ML on muscle repair after injury. The response of injured muscle to robot-actuated cyclic compressive loading was found to be age sensitive, revealing not only a lack of reparative benefit of ML on injured aged muscles but also exacerbation of tissue inflammation. ML alone also disrupted the normal regenerative processes of aged muscle stem cells. However, these negative effects could be reversed by introducing anti-inflammatory therapy alongside ML application, leading to enhanced skeletal muscle regeneration even in aged mice.
Collapse
Affiliation(s)
- S L McNamara
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - B R Seo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - B R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - E B Roloson
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - J T Alvarez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - C T O'Neill
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - H H Vandenburgh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - C J Walsh
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - D J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| |
Collapse
|
12
|
Lian J, Walker RG, D'Amico A, Vujic A, Mills MJ, Messemer KA, Mendello KR, Goldstein JM, Leacock KA, Epp S, Stimpfl EV, Thompson TB, Wagers AJ, Lee RT. Functional substitutions of amino acids that differ between GDF11 and GDF8 impact skeletal development and skeletal muscle. Life Sci Alliance 2023; 6:e202201662. [PMID: 36631218 PMCID: PMC9834663 DOI: 10.26508/lsa.202201662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
Growth differentiation factor 11 (GDF11) and GDF8 (MSTN) are closely related TGF-β family proteins that interact with nearly identical signaling receptors and antagonists. However, GDF11 appears to activate SMAD2/3 more potently than GDF8 in vitro and in vivo. The ligands possess divergent structural properties, whereby substituting unique GDF11 amino acids into GDF8 enhanced the activity of the resulting chimeric GDF8. We investigated potentially distinct endogenous activities of GDF11 and GDF8 in vivo by genetically modifying their mature signaling domains. Full recoding of GDF8 to that of GDF11 yielded mice lacking GDF8, with GDF11 levels ∼50-fold higher than normal, and exhibiting modestly decreased muscle mass, with no apparent negative impacts on health or survival. Substitution of two specific amino acids in the fingertip region of GDF11 with the corresponding GDF8 residues resulted in prenatal axial skeletal transformations, consistent with Gdf11-deficient mice, without apparent perturbation of skeletal or cardiac muscle development or homeostasis. These experiments uncover distinctive features between the GDF11 and GDF8 mature domains in vivo and identify a specific requirement for GDF11 in early-stage skeletal development.
Collapse
Affiliation(s)
- John Lian
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ryan G Walker
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Andrea D'Amico
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Melanie J Mills
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kathleen A Messemer
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kourtney R Mendello
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Jill M Goldstein
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Krystynne A Leacock
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Soraya Epp
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Emma V Stimpfl
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, OH, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Joslin Diabetes Center, Boston, MA, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
13
|
Lazure F, Farouni R, Sahinyan K, Blackburn DM, Hernández-Corchado A, Perron G, Lu T, Osakwe A, Ragoussis J, Crist C, Perkins TJ, Jahani-Asl A, Najafabadi HS, Soleimani VD. Transcriptional reprogramming of skeletal muscle stem cells by the niche environment. Nat Commun 2023; 14:535. [PMID: 36726011 PMCID: PMC9892560 DOI: 10.1038/s41467-023-36265-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Adult stem cells are indispensable for tissue regeneration, but their function declines with age. The niche environment in which the stem cells reside plays a critical role in their function. However, quantification of the niche effect on stem cell function is lacking. Using muscle stem cells (MuSC) as a model, we show that aging leads to a significant transcriptomic shift in their subpopulations accompanied by locus-specific gain and loss of chromatin accessibility and DNA methylation. By combining in vivo MuSC transplantation and computational methods, we show that the expression of approximately half of all age-altered genes in MuSCs from aged male mice can be restored by exposure to a young niche environment. While there is a correlation between gene reversibility and epigenetic alterations, restoration of gene expression occurs primarily at the level of transcription. The stem cell niche environment therefore represents an important therapeutic target to enhance tissue regeneration in aging.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Rick Farouni
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Korin Sahinyan
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Aldo Hernández-Corchado
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Gabrielle Perron
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.,Quantitative Life Sciences, McGill University, Montreal, Canada
| | - Adrien Osakwe
- Quantitative Life Sciences, McGill University, Montreal, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Colin Crist
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine and University of Ottawa Brain and Mind Research Institute, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Hamed S Najafabadi
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada. .,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada. .,Quantitative Life Sciences, McGill University, Montreal, Canada.
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada. .,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.
| |
Collapse
|
14
|
Millozzi F, Papait A, Bouché M, Parolini O, Palacios D. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging? Int J Mol Sci 2023; 24:1175. [PMID: 36674691 PMCID: PMC9862642 DOI: 10.3390/ijms24021175] [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: 11/07/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
The skeletal muscle has a very remarkable ability to regenerate upon injury under physiological conditions; however, this regenerative capacity is strongly diminished in physio-pathological conditions, such as those present in diseased or aged muscles. Many muscular dystrophies (MDs) are characterized by aberrant inflammation due to the deregulation of both the lymphoid and myeloid cell populations and the production of pro-inflammatory cytokines. Pathological inflammation is also observed in old muscles due to a systemic change in the immune system, known as "inflammaging". Immunomodulation represents, therefore, a promising therapeutic opportunity for different skeletal muscle conditions. However, the use of immunomodulatory drugs in the clinics presents several caveats, including their low stability in vivo, the need for high doses to obtain therapeutically relevant effects, and the presence of strong side effects. Within this context, the emerging field of nanomedicine provides the powerful tools needed to control the immune response. Nano-scale materials are currently being explored as biocarriers to release immunomodulatory agents in the damaged tissues, allowing therapeutic doses with limited off-target effects. In addition, the intrinsic immunomodulatory properties of some nanomaterials offer further opportunities for intervention that still need to be systematically explored. Here we exhaustively review the state-of-the-art regarding the use of nano-sized materials to modulate the aberrant immune response that characterizes some physio-pathological muscle conditions, such as MDs or sarcopenia (the age-dependent loss of muscle mass). Based on our learnings from cancer and immune tolerance induction, we also discuss further opportunities, challenges, and limitations of the emerging field of nano-immunomodulation.
Collapse
Affiliation(s)
- Francesco Millozzi
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Andrea Papait
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Marina Bouché
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| |
Collapse
|
15
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
16
|
Kolodziej F, McDonagh B, Burns N, Goljanek-Whysall K. MicroRNAs as the Sentinels of Redox and Hypertrophic Signalling. Int J Mol Sci 2022; 23:ijms232314716. [PMID: 36499053 PMCID: PMC9737617 DOI: 10.3390/ijms232314716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
Oxidative stress and inflammation are associated with skeletal muscle function decline with ageing or disease or inadequate exercise and/or poor diet. Paradoxically, reactive oxygen species and inflammatory cytokines are key for mounting the muscular and systemic adaptive responses to endurance and resistance exercise. Both ageing and lifestyle-related metabolic dysfunction are strongly linked to exercise redox and hypertrophic insensitivity. The adaptive inability and consequent exercise intolerance may discourage people from physical training resulting in a vicious cycle of under-exercising, energy surplus, chronic mitochondrial stress, accelerated functional decline and increased susceptibility to serious diseases. Skeletal muscles are malleable and dynamic organs, rewiring their metabolism depending on the metabolic or mechanical stress resulting in a specific phenotype. Endogenous RNA silencing molecules, microRNAs, are regulators of these metabolic/phenotypic shifts in skeletal muscles. Skeletal muscle microRNA profiles at baseline and in response to exercise have been observed to differ between adult and older people, as well as trained vs. sedentary individuals. Likewise, the circulating microRNA blueprint varies based on age and training status. Therefore, microRNAs emerge as key regulators of metabolic health/capacity and hormetic adaptability. In this narrative review, we summarise the literature exploring the links between microRNAs and skeletal muscle, as well as systemic adaptation to exercise. We expand a mathematical model of microRNA burst during adaptation to exercise through supporting data from the literature. We describe a potential link between the microRNA-dependent regulation of redox-signalling sensitivity and the ability to mount a hypertrophic response to exercise or nutritional cues. We propose a hypothetical model of endurance exercise-induced microRNA "memory cloud" responsible for establishing a landscape conducive to aerobic as well as anabolic adaptation. We suggest that regular aerobic exercise, complimented by a healthy diet, in addition to promoting mitochondrial health and hypertrophic/insulin sensitivity, may also suppress the glycolytic phenotype and mTOR signalling through miRNAs which in turn promote systemic metabolic health.
Collapse
Affiliation(s)
- Filip Kolodziej
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Brian McDonagh
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Nicole Burns
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Katarzyna Goljanek-Whysall
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
- Institute of Life Course and Medical Science, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
17
|
Liang Z, Zhang T, Liu H, Li Z, Peng L, Wang C, Wang T. Inflammaging: The ground for sarcopenia? Exp Gerontol 2022; 168:111931. [PMID: 35985553 DOI: 10.1016/j.exger.2022.111931] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022]
Abstract
Sarcopenia is a progressive skeletal muscle disease that occurs most commonly in the elderly population, contributing to increased costs and hospitalization. Exercise and nutritional therapy have been proven to be effective for sarcopenia, and some drugs can also alleviate declines in muscle mass and function due to sarcopenia. However, there is no specific pharmacological treatment for sarcopenia at present. This review will mainly discuss the relationship between inflammaging and sarcopenia. The increased secretion of proinflammatory cytokines with aging may be because of cellular senescence, immunosenescence, alterations in adipose tissue, damage-associated molecular patterns (DAMPs), and gut microbes due to aging. These sources of inflammaging can impact the sarcopenia process through direct or indirect pathways. Conversely, sarcopenia can also aggravate the process of inflammaging, creating a vicious cycle. Targeting sources of inflammaging can influence muscle function, which could be considered a therapeutic target for sarcopenia. Moreover, not only proinflammatory cytokines but also anti-inflammatory cytokines can influence muscle and inflammation and participate in the progression of sarcopenia. This review focuses on the effects of TNF-α, IL-6, and IL-10, which can be detected in plasma. Therefore, clearing chronic inflammation by targeting proinflammatory cytokines (TNF-α, IL-1, IL-6) and the inflammatory pathway (JAK/STAT, autophagy, NF-κB) may be effective in treating sarcopenia.
Collapse
Affiliation(s)
- Zejun Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tianxiao Zhang
- School of Healthcare Sciences, Cardiff University, Health Park, CF14 4XN Wales, UK
| | - Honghong Liu
- West China School of Nursing/West China Hospital, Sichuan University, NO.37 Alley, Chengdu 610041, Sichuan, PR China
| | - Zhenlin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lihong Peng
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, PR China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
18
|
Huo F, Liu Q, Liu H. Contribution of muscle satellite cells to sarcopenia. Front Physiol 2022; 13:892749. [PMID: 36035464 PMCID: PMC9411786 DOI: 10.3389/fphys.2022.892749] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Sarcopenia, a disorder characterized by age-related muscle loss and reduced muscle strength, is associated with decreased individual independence and quality of life, as well as a high risk of death. Skeletal muscle houses a normally mitotically quiescent population of adult stem cells called muscle satellite cells (MuSCs) that are responsible for muscle maintenance, growth, repair, and regeneration throughout the life cycle. Patients with sarcopenia are often exhibit dysregulation of MuSCs homeostasis. In this review, we focus on the etiology, assessment, and treatment of sarcopenia. We also discuss phenotypic and regulatory mechanisms of MuSC quiescence, activation, and aging states, as well as the controversy between MuSC depletion and sarcopenia. Finally, we give a multi-dimensional treatment strategy for sarcopenia based on improving MuSC function.
Collapse
Affiliation(s)
- Fengjiao Huo
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, China
- *Correspondence: Hailiang Liu,
| |
Collapse
|
19
|
Hong X, Campanario S, Ramírez-Pardo I, Grima-Terrén M, Isern J, Muñoz-Cánoves P. Stem cell aging in the skeletal muscle: The importance of communication. Ageing Res Rev 2022; 73:101528. [PMID: 34818593 DOI: 10.1016/j.arr.2021.101528] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 01/04/2023]
Abstract
Adult stem cells sustain tissue homeostasis and regeneration; their functional decline is often linked to aging, which is characterized by the progressive loss of physiological functions across multiple tissues and organs. The resident stem cells in skeletal muscle, termed satellite cells, are normally quiescent but activate upon injury to reconstitute the damaged tissue. In this review, we discuss the current understanding of the molecular processes that contribute to the functional failure of satellite cells during aging. This failure is due not only to intrinsic changes but also to extrinsic factors, most of which are still undefined but originate from the muscle tissue microenvironment of the satellite cells (the niche), or from the systemic environment. We also highlight the emerging applications of the powerful single-cell sequencing technologies in the study of skeletal muscle aging, particularly in the heterogeneity of the satellite cell population and the molecular interaction of satellite cells and other cell types in the niche. An improved understanding of how satellite cells communicate with their environment, and how this communication is perturbed with aging, will be helpful for defining countermeasures against loss of muscle regenerative capacity in sarcopenia.
Collapse
Affiliation(s)
- Xiaotong Hong
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain
| | - Silvia Campanario
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain
| | - Ignacio Ramírez-Pardo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain
| | - Mercedes Grima-Terrén
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain
| | - Joan Isern
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), E-28029 Madrid, Spain; Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), E-08003 Barcelona, Spain; ICREA, E-08010 Barcelona, Spain.
| |
Collapse
|
20
|
Orkaby AR, Dufour AB, Yang L, Sesso HD, Gaziano JM, Djousse L, Driver JA, Travison TG. Long-Term Aspirin Use and Self-Reported Walking Speed in Older Men: The Physicians' Health Study. J Frailty Aging 2022; 11:12-17. [PMID: 35122085 PMCID: PMC8818085 DOI: 10.14283/jfa.2021.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Mobility limitation is a component of frailty that shares a bidirectional relationship with cardiovascular disease (CVD). Data are limited on the role of established CVD prevention therapies, such as aspirin, for prevention of frailty and mobility limitation. OBJECTIVES Examine the association between long-term aspirin use and walking speed. DESIGN, SETTING, PARTICIPANTS Prospective cohort of 14,315 men who participated in the Physicians' Health Study I, a completed randomized controlled trial of aspirin (1982-1988), with extended post-trial follow-up. MEASUREMENTS Annual questionnaires collected data on aspirin use, lifestyle and other factors. Average annual aspirin use was categorized for each participant: ≤60 days/year and >60 days/year. Mobility was defined according to self-reported walking pace, categorized as: don't walk regularly (reference), easy/casual <2mph, normal ≥2-2.9mph, or brisk/very brisk ≥3mph. Propensity scoring balanced covariates between aspirin categories. Multinomial logistic regression models estimated odds of being in each self-reported walking category. RESULTS Mean age was 70±8 years; mean aspirin use 11 years. There were 2,056 (14.3%) participants who reported aspirin use ≤60 days/year. Aspirin use >60 days/year was associated with drinking alcohol, smoking, hypertension, heart disease and stroke, while ≤60 days/year was associated with anticoagulation use and bleeding history. In all, 13% reported not walking regularly, 12% walked <2 mph, 44% walked ≥2-2.9 mph, and 31% walked ≥3 mph. After propensity score adjustment, regular aspirin use was associated with a faster walking speed. Odds ratios (95% confidence intervals) were 1.16 (0.97 to 1.39), 1.24 (1.08 to 1.43), and 1.40 (1.21 to 1.63) for <2 mph, ≥2-2.9 mph and ≥3 mph, respectively, compared to not walking regularly (p-trend<0.001). CONCLUSIONS In this cohort of older men, long-term aspirin use is associated with a greater probability of faster walking speed later in life.
Collapse
Affiliation(s)
- Ariela R. Orkaby
- New England GRECC, VA Boston Healthcare System, Boston, MA;,Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - Alyssa B. Dufour
- Marcus Institute for Aging Research, Hebrew Senior Life, and Harvard Medical School, Boston, MA
| | - Laiji Yang
- Marcus Institute for Aging Research, Hebrew Senior Life, and Harvard Medical School, Boston, MA
| | - Howard D. Sesso
- Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - J. Michael Gaziano
- New England GRECC, VA Boston Healthcare System, Boston, MA;,Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - Luc Djousse
- New England GRECC, VA Boston Healthcare System, Boston, MA;,Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - Jane A. Driver
- New England GRECC, VA Boston Healthcare System, Boston, MA;,Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - Thomas G. Travison
- Marcus Institute for Aging Research, Hebrew Senior Life, and Harvard Medical School, Boston, MA
| |
Collapse
|
21
|
Abrigo J, Simon F, Cabrera D, Vilos C, Cabello-Verrugio C. Combined Administration of Andrographolide and Angiotensin- (1-7) Synergically Increases the Muscle Function and Strength in Aged Mice. Curr Mol Med 2021; 22:908-918. [PMID: 34875988 DOI: 10.2174/1566524021666211207112106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Sarcopenia is a progressive and generalized skeletal muscle disorder characterized by muscle weakness, loss of muscle mass, and decline in the capacity of force generation. Aging can cause sarcopenia. Several therapeutic strategies have been evaluated to prevent or alleviate this disorder. One of them is angiotensin 1-7 [Ang-(1-7)], an anti-atrophic peptide for skeletal muscles that regulates decreased muscle mass for several causes, including aging. Another regulator of muscle mass and function is andrographolide, a bicyclic diterpenoid lactone that decreases the nuclear factor kappa B (NF-κB) signaling and attenuates the severity of some muscle diseases. OBJECTIVE Evaluate the effect of combined administration of Ang-(1-7) with andrographolide on the physical performance, muscle strength, and fiber´s diameter in a murine model of sarcopenia by aging. METHODS Aged male mice of the C57BL/6J strain were treated with Andrographolide, Ang-(1-7), or combined for three months. The physical performance, muscle strength, and fiber´s diameter were measured. RESULTS The results showed that aged mice (24 months old) treated with Ang-(1-7) or Andrographolide improved their performance on a treadmill test, muscle strength, and their fiber´s diameter compared to aged mice without treatment. The combined administration of Ang-(1-7) with andrographolide to aged mice has an enhanced synergically effect on physical performance, muscle strength, and fiber´s diameter. CONCLUSION Our results indicated that in aged mice, the effects of andrographolide and Ang-(1-7) on muscle function, strength, and fiber´s diameter are potentiated.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello. Santiago. Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy. Santiago. Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile. Santiago. Chile
| | - Cristian Vilos
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile. Santiago. Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello. Santiago. Chile
| |
Collapse
|
22
|
Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 2021; 23:204-226. [PMID: 34663964 DOI: 10.1038/s41580-021-00421-2] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle contains a designated population of adult stem cells, called satellite cells, which are generally quiescent. In homeostasis, satellite cells proliferate only sporadically and usually by asymmetric cell division to replace myofibres damaged by daily activity and maintain the stem cell pool. However, satellite cells can also be robustly activated upon tissue injury, after which they undergo symmetric divisions to generate new stem cells and numerous proliferating myoblasts that later differentiate to muscle cells (myocytes) to rebuild the muscle fibre, thereby supporting skeletal muscle regeneration. Recent discoveries show that satellite cells have a great degree of population heterogeneity, and that their cell fate choices during the regeneration process are dictated by both intrinsic and extrinsic mechanisms. Extrinsic cues come largely from communication with the numerous distinct stromal cell types in their niche, creating a dynamically interactive microenvironment. This Review discusses the role and regulation of satellite cells in skeletal muscle homeostasis and regeneration. In particular, we highlight the cell-intrinsic control of quiescence versus activation, the importance of satellite cell-niche communication, and deregulation of these mechanisms associated with ageing. The increasing understanding of how satellite cells are regulated will help to advance muscle regeneration and rejuvenation therapies.
Collapse
|
23
|
Yuan S, Zheng S, Zheng K, Gao Y, Chen M, Li Y, Bai X. Sympathetic activity is correlated with satellite cell aging and myogenesis via β2-adrenoceptor. Stem Cell Res Ther 2021; 12:505. [PMID: 34530910 PMCID: PMC8447727 DOI: 10.1186/s13287-021-02571-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/26/2021] [Indexed: 02/09/2023] Open
Abstract
Background and objective Sympathetic activity plays an important role in the proliferation and differentiation of stem cells, and it changes over time, thereby exerting differential effects on various stem cell types. Aging causes sympathetic hyperactivity in aged tissues and blunts sympathetic nerves regulation, and sympathetic abnormalities play a role in aging-related diseases. Currently, the effect of sympathetic activity on skeletal muscle stem cells, namely satellite cells (SCs), is unclear. This study aimed to investigate the effects of skeletal muscle sympathetic activity on SC aging and skeletal muscle repair. Materials and methods To evaluate skeletal muscle and fibrotic areas, numbers of SCs and myonuclei per muscle fiber, β2-adrenoceptor (β2-ADR) expression, muscle repair, and sympathetic innervation in skeletal muscle, aged mice, young mice that underwent chemical sympathectomy (CS) were utilized. Mice with a tibialis anterior muscle injury were treated by barium chloride (BaCl2) and clenbuterol (CLB) in vivo. SCs or C2C12 cells were differentiated into myotubes and treated with or without CLB. Immunofluorescence, western blot, sirius red, and hematoxylin–eosin were used to evaluate SCs, myogenic repair and differentiation. Results The number of SCs, sympathetic activity, and reparability of muscle injury in aged mice were significantly decreased, compared with those in young mice. The above characteristics of young mice that underwent CS were similar to those of aged mice. While CLB promoted the repair of muscle injury in aged and CS mice and ameliorated the reduction in the SC number and sympathetic activity, the effects of CLB on the SCs and sympathetic nerves in young mice were not significant. CLB inhibited the myogenic differentiation of C2C12 cells in vitro. We further found that NF-κB and ERK1/2 signaling pathways were activated during myogenic differentiation, and this process could be inhibited by CLB. Conclusion Normal sympathetic activity promoted the stemness of SCs to thereby maintain a steady state. It also could maintain total and self-renewing number of SCs and maintain a quiescent state, which was correlated with skeletal SCs via β2-ADR. Normal sympathetic activity was also beneficial for the myogenic repair of injured skeletal muscle. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02571-8.
Collapse
Affiliation(s)
- Shiguo Yuan
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China.,Department of Orthopaedic Surgery, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, 570203, China
| | - Sheng Zheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kai Zheng
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yanping Gao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Meixiong Chen
- Department of Orthopaedic Surgery, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, 570203, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
24
|
Opportunities and Challenges in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1341:143-175. [PMID: 33748933 DOI: 10.1007/5584_2021_624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studying aging, as a physiological process that can cause various pathological phenotypes, has attracted lots of attention due to its increasing burden and prevalence. Therefore, understanding its mechanism to find novel therapeutic alternatives for age-related disorders such as neurodegenerative and cardiovascular diseases is essential. Stem cell senescence plays an important role in aging. In the context of the underlying pathways, mitochondrial dysfunction, epigenetic and genetic alterations, and other mechanisms have been studied and as a consequence, several rejuvenation strategies targeting these mechanisms like pharmaceutical interventions, genetic modification, and cellular reprogramming have been proposed. On the other hand, since stem cells have great potential for disease modeling, they have been useful for representing aging and its associated disorders. Accordingly, the main mechanisms of senescence in stem cells and promising ways of rejuvenation, along with some examples of stem cell models for aging are introduced and discussed. This review aims to prepare a comprehensive summary of the findings by focusing on the most recent ones to shine a light on this area of research.
Collapse
|
25
|
Vennekens A, Laporte E, Hermans F, Cox B, Modave E, Janiszewski A, Nys C, Kobayashi H, Malengier-Devlies B, Chappell J, Matthys P, Garcia MI, Pasque V, Lambrechts D, Vankelecom H. Interleukin-6 is an activator of pituitary stem cells upon local damage, a competence quenched in the aging gland. Proc Natl Acad Sci U S A 2021; 118:e2100052118. [PMID: 34161279 PMCID: PMC8237615 DOI: 10.1073/pnas.2100052118] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Stem cells in the adult pituitary are quiescent yet show acute activation upon tissue injury. The molecular mechanisms underlying this reaction are completely unknown. We applied single-cell transcriptomics to start unraveling the acute pituitary stem cell activation process as occurring upon targeted endocrine cell-ablation damage. This stem cell reaction was contrasted with the aging (middle-aged) pituitary, known to have lost damage-repair capacity. Stem cells in the aging pituitary show regressed proliferative activation upon injury and diminished in vitro organoid formation. Single-cell RNA sequencing uncovered interleukin-6 (IL-6) as being up-regulated upon damage, however only in young but not aging pituitary. Administering IL-6 to young mice promptly triggered pituitary stem cell proliferation, while blocking IL-6 or associated signaling pathways inhibited such reaction to damage. By contrast, IL-6 did not generate a pituitary stem cell activation response in aging mice, coinciding with elevated basal IL-6 levels and raised inflammatory state in the aging gland (inflammaging). Intriguingly, in vitro stem cell activation by IL-6 was discerned in organoid culture not only from young but also from aging pituitary, indicating that the aging gland's stem cells retain intrinsic activatability in vivo, likely impeded by the prevailing inflammatory tissue milieu. Importantly, IL-6 supplementation strongly enhanced the growth capability of pituitary stem cell organoids, thereby expanding their potential as an experimental model. Our study identifies IL-6 as a pituitary stem cell activator upon local damage, a competence quenched at aging, concomitant with raised IL-6/inflammatory levels in the older gland. These insights may open the way to interfering with pituitary aging.
Collapse
Affiliation(s)
- Annelies Vennekens
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Emma Laporte
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Florian Hermans
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Laboratory of Morphology, Biomedical Research Institute, Hasselt University, 3590 Diepenbeek, Belgium
| | - Benoit Cox
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Elodie Modave
- Center for Cancer Biology, Vlaams Instituut voor Biotechnologie, 3000 Leuven, Belgium
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Adrian Janiszewski
- Laboratory for Cellular Reprogramming and Epigenetic Regulation, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Charlotte Nys
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Hiroto Kobayashi
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Department of Anatomy and Structural Science, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| | - Bert Malengier-Devlies
- Immunity and Inflammation Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Joel Chappell
- Laboratory for Cellular Reprogramming and Epigenetic Regulation, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Patrick Matthys
- Immunity and Inflammation Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Marie-Isabelle Garcia
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Faculty of Medicine, Université Libre de Bruxelles, 1070 Bruxelles, Belgium
| | - Vincent Pasque
- Laboratory for Cellular Reprogramming and Epigenetic Regulation, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Diether Lambrechts
- Center for Cancer Biology, Vlaams Instituut voor Biotechnologie, 3000 Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
26
|
Endo Y, Karvar M, Sinha I. Muscle Cryoinjury and Quantification of Regenerating Myofibers in Mice. Bio Protoc 2021; 11:e4036. [PMID: 34250203 DOI: 10.21769/bioprotoc.4036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/02/2021] [Accepted: 04/23/2021] [Indexed: 11/02/2022] Open
Abstract
Cryoinjury, or injury due to freezing, is a method of creating reproducible, local injuries in skeletal muscle. This method allows studying the regenerative response following muscle injuries in vivo, thus enabling the evaluation of local and systemic factors that influence the processes of myofiber regeneration. Cryoinjuries are applicable to the study of various modalities of muscle injury, particularly non-traumatic and traumatic injuries, without a loss of substantial volume of muscle mass. Cryoinjury requires only simple instruments and has the advantage over other methods that the extent of the lesion can be easily adjusted and standardized according to the duration of contact with the freezing instrument. The regenerative response can be evaluated histologically by the average maturity of regenerating myofibers as indicated by the cross-sectional areas of myofibers with centrally located nuclei. Accordingly, cryoinjury is regarded as one of the most reliable and easily accessible methods for simulating muscle injuries in studies of muscle regeneration.
Collapse
Affiliation(s)
- Yori Endo
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Mehran Karvar
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Indranil Sinha
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Harvard Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
27
|
Orkaby AR, Yang L, Dufour AB, Travison TG, Sesso HD, Driver JA, Djousse L, Gaziano JM. Association Between Long-Term Aspirin Use and Frailty in Men: The Physicians' Health Study. J Gerontol A Biol Sci Med Sci 2021; 76:1077-1083. [PMID: 32918079 DOI: 10.1093/gerona/glaa233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Chronic inflammation may lead to frailty, however the potential for anti-inflammatory medications such as aspirin to prevent frailty is unknown. We sought to examine the association between long-term aspirin use and prevalent frailty. METHODS We included 12 101 men ≥60 years who participated in the Physicians' Health Study I, a completed aspirin randomized controlled trial (1982-1989). Annual questionnaires collected self-reported data on daily aspirin use, lifestyle, and clinical variables. Average aspirin use was summed into 2 categories: ≤60 days/year and >60 days/year. Frailty was assessed using a 33-item index 11 years after trial completion. A score of ≥0.21 was considered frail. Propensity score inverse probability of treatment weighting was used for statistical control of confounding. Logistic regression models estimated odds of frailty as a function of categories of average aspirin use. RESULTS Mean age was 70.5 years (range 60-101). Following an average of 11 ± 0.6 years of follow-up, aspirin use was reported as ≤60 days/year for 15%; 2413 participants (20%) were frail. Frequency of aspirin use was associated with smoking, alcohol consumption, hypertension, and cardiovascular disease, but negatively associated with bleeding and Coumadin use. The odds ratio (95% confidence intervals) for frailty was 0.85 (0.76-0.96) for average aspirin use >60 days/year versus aspirin use ≤60 days/year. Results were similar using an alternate definition of frailty. CONCLUSIONS Long-term regular aspirin use is inversely associated with frailty among older men, even after consideration of multimorbidity and health behaviors. Work is needed to understand the role of medications with anti-inflammatory properties on aging.
Collapse
Affiliation(s)
- Ariela R Orkaby
- New England GRECC (Geriatric Research, Education, and Clinical Center), VA Boston Healthcare System, Massachusetts.,Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Laiji Yang
- Institute for Aging Research, Hebrew Senior Life, Harvard Medical School, Boston, Massachusetts
| | - Alyssa B Dufour
- Institute for Aging Research, Hebrew Senior Life, Harvard Medical School, Boston, Massachusetts
| | - Thomas G Travison
- Institute for Aging Research, Hebrew Senior Life, Harvard Medical School, Boston, Massachusetts
| | - Howard D Sesso
- Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Preventive Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jane A Driver
- New England GRECC (Geriatric Research, Education, and Clinical Center), VA Boston Healthcare System, Massachusetts.,Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Luc Djousse
- Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts.,MAVERIC (Massachusetts Veterans Epidemiology Research and Information Center), VA Boston Healthcare System, Massachusetts
| | - J Michael Gaziano
- Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts.,MAVERIC (Massachusetts Veterans Epidemiology Research and Information Center), VA Boston Healthcare System, Massachusetts
| |
Collapse
|
28
|
Tichy ED, Ma N, Sidibe D, Loro E, Kocan J, Chen DZ, Khurana TS, Hasty P, Mourkioti F. Persistent NF-κB activation in muscle stem cells induces proliferation-independent telomere shortening. Cell Rep 2021; 35:109098. [PMID: 33979621 PMCID: PMC8183356 DOI: 10.1016/j.celrep.2021.109098] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/30/2020] [Accepted: 04/16/2021] [Indexed: 12/30/2022] Open
Abstract
During the repeated cycles of damage and repair in many muscle disorders, including Duchenne muscular dystrophy (DMD), the muscle stem cell (MuSC) pool becomes less efficient at responding to and repairing damage. The underlying mechanism of such stem cell dysfunction is not fully known. Here, we demonstrate that the distinct early telomere shortening of diseased MuSCs in both mice and young DMD patients is associated with aberrant NF-κB activation. We find that prolonged NF-κB activation in MuSCs in chronic injuries leads to shortened telomeres and Ku80 dysregulation and results in severe skeletal muscle defects. Our studies provide evidence of a role for NF-κB in regulating stem-cell-specific telomere length, independently of cell replication, and could be a congruent mechanism that is applicable to additional tissues and/or diseases characterized by systemic chronic inflammation. Tichy et al. reveal a role for NF-κB signaling in regulating telomere length in muscle stem cells (MuSCs) after chronic injuries. Persistent activation of NF-κB leads to shortened telomeres, Ku80 dysregulation, and muscle defects. The findings link stem cell dysfunction and NF-κB-dependent telomere shortening in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Sidibe
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emanuele Loro
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Kocan
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delia Z Chen
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tejvir S Khurana
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Hasty
- The Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Regenerative Medicine, Musculoskeletal Regeneration Program, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Endo Y, Baldino K, Li B, Zhang Y, Sakthivel D, MacArthur M, Panayi AC, Kip P, Spencer DJ, Jasuja R, Bagchi D, Bhasin S, Nuutila K, Neppl RL, Wagers AJ, Sinha I. Loss of ARNT in skeletal muscle limits muscle regeneration in aging. FASEB J 2020; 34:16086-16104. [PMID: 33064329 PMCID: PMC7756517 DOI: 10.1096/fj.202000761rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
The ability of skeletal muscle to regenerate declines significantly with aging. The expression of aryl hydrocarbon receptor nuclear translocator (ARNT), a critical component of the hypoxia signaling pathway, was less abundant in skeletal muscle of old (23-25 months old) mice. This loss of ARNT was associated with decreased levels of Notch1 intracellular domain (N1ICD) and impaired regenerative response to injury in comparison to young (2-3 months old) mice. Knockdown of ARNT in a primary muscle cell line impaired differentiation in vitro. Skeletal muscle-specific ARNT deletion in young mice resulted in decreased levels of whole muscle N1ICD and limited muscle regeneration. Administration of a systemic hypoxia pathway activator (ML228), which simulates the actions of ARNT, rescued skeletal muscle regeneration in both old and ARNT-deleted mice. These results suggest that the loss of ARNT in skeletal muscle is partially responsible for diminished myogenic potential in aging and activation of hypoxia signaling holds promise for rescuing regenerative activity in old muscle.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Kodi Baldino
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Bin Li
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuteng Zhang
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | | | - Michael MacArthur
- Department of Genetics and Complex DiseasesHarvard School of Public HealthBostonMAUSA
- Division of Vascular and Endovascular SurgeryBrigham and Women's HospitalBostonMAUSA
| | - Adriana C. Panayi
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Peter Kip
- Division of Vascular and Endovascular SurgeryBrigham and Women's HospitalBostonMAUSA
| | | | - Ravi Jasuja
- Division of EndocrinologyBrigham and Women's HospitalBostonMAUSA
| | - Debalina Bagchi
- Department of Orthopedic SurgeryBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Shalender Bhasin
- Division of EndocrinologyBrigham and Women's HospitalBostonMAUSA
| | - Kristo Nuutila
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Ronald L. Neppl
- Department of Orthopedic SurgeryBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Amy J. Wagers
- Joslin Diabetes CenterBostonMAUSA
- Harvard Department of Stem Cell and Regenerative BiologyHarvard Stem Cell InstituteCambridgeMAUSA
- Paul F. Glenn Center for the Biology of AgingHarvard Medical SchoolBostonMAUSA
| | - Indranil Sinha
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Harvard Department of Stem Cell and Regenerative BiologyHarvard Stem Cell InstituteCambridgeMAUSA
| |
Collapse
|
30
|
Wang H, Wang H, Li X, Xu W. Characteristics of Early Internal Laryngeal Muscle Atrophy After Recurrent Laryngeal Nerve Injuries in Rats. Laryngoscope 2020; 131:E1256-E1264. [PMID: 33098577 DOI: 10.1002/lary.29210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 11/10/2022]
Abstract
OBJECTIVES/HYPOTHESIS The present study investigated the characteristics of early internal laryngeal muscle atrophy in recurrent laryngeal nerve injury (RLNI) rats. STUDY DESIGN To observe the characteristics of early internal laryngeal muscle atrophy post RLNI. METHODS Rats were divided into three groups: sham-operated control group (n = 20), recurrent laryngeal nerve transverse injury group (RLNTI, n = 50), and recurrent laryngeal nerve blunt contusion group (RLNBC, n = 50). Five weeks after RLNI, certain rats were sacrificed weekly, and their laryngeal tissues were harvested. The atrophic features of internal laryngeal muscles were detected using hematoxylin and eosin. NF-κB and MuRF-1 levels were tested using IHC. RESULTS The atrophic degree and fibrosis of thyroarytenoid, posterior cricoarytenoid, and lateral cricoarytenoid muscles were related to the type of RLNI. The average myofiber cross-sectional areas increased before an obvious decrease in the RLNTI and RLNBC groups. Muscle recovery occurred in the RLNBC group starting 4 weeks after RLNI, but only a weak trend was observed in the RLNTI group in the 5th week. During the muscle atrophy process, MuRF-1 and NF-κB were upregulated early and were maintained at a high level, which showed a trend similar to muscle atrophy. However, NF-κB expression was opposite to MuRF-1 expression and muscle atrophy when the muscles recovered. CONCLUSION The atrophy degree of internal laryngeal muscles was associated with the type of RLNI. The NF-κB/MuRF-1 signaling pathway was involved in internal laryngeal muscle atrophy after RLNI, which is different from skeletal muscle after denervation. LEVEL OF EVIDENCE NA Laryngoscope, 131:E1256-E1264, 2021.
Collapse
Affiliation(s)
- Hong Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Haizhou Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Xueyan Li
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Wen Xu
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Endo Y, Nourmahnad A, Sinha I. Optimizing Skeletal Muscle Anabolic Response to Resistance Training in Aging. Front Physiol 2020; 11:874. [PMID: 32792984 PMCID: PMC7390896 DOI: 10.3389/fphys.2020.00874] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Loss of muscle mass and strength with aging, also termed sarcopenia, results in a loss of mobility and independence. Exercise, particularly resistance training, has proven to be beneficial in counteracting the aging-associated loss of skeletal muscle mass and function. However, the anabolic response to exercise in old age is not as robust, with blunted improvements in muscle size, strength, and function in comparison to younger individuals. This review provides an overview of several physiological changes which may contribute to age-related loss of muscle mass and decreased anabolism in response to resistance training in the elderly. Additionally, the following supplemental therapies with potential to synergize with resistance training to increase muscle mass are discussed: nutrition, creatine, anti-inflammatory drugs, testosterone, and growth hormone (GH). Although these interventions hold some promise, further research is necessary to optimize the response to exercise in elderly patients.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Atousa Nourmahnad
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, United States
| |
Collapse
|
32
|
Straughn AR, Hindi SM, Xiong G, Kumar A. Canonical NF-κB signaling regulates satellite stem cell homeostasis and function during regenerative myogenesis. J Mol Cell Biol 2020; 11:53-66. [PMID: 30239789 DOI: 10.1093/jmcb/mjy053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/18/2018] [Indexed: 01/08/2023] Open
Abstract
Skeletal muscle regeneration in adults is attributed to the presence of satellite stem cells that proliferate, differentiate, and eventually fuse with injured myofibers. However, the signaling mechanisms that regulate satellite cell homeostasis and function remain less understood. While IKKβ-mediated canonical NF-κB signaling has been implicated in the regulation of myogenesis and skeletal muscle mass, its role in the regulation of satellite cell function during muscle regeneration has not been fully elucidated. Here, we report that canonical NF-κB signaling is induced in skeletal muscle upon injury. Satellite cell-specific inducible ablation of IKKβ attenuates skeletal muscle regeneration in adult mice. Targeted ablation of IKKβ also reduces the number of satellite cells in injured skeletal muscle of adult mice, potentially through inhibiting their proliferation and survival. We also demonstrate that the inhibition of specific components of the canonical NF-κB pathway causes precocious differentiation of cultured satellite cells both ex vivo and in vitro. Finally, our results highlight that the constitutive activation of canonical NF-κB signaling in satellite cells also attenuates skeletal muscle regeneration following injury in adult mice. Collectively, our study demonstrates that the proper regulation of canonical NF-κB signaling is important for the regeneration of adult skeletal muscle.
Collapse
Affiliation(s)
- Alex R Straughn
- Departments of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Sajedah M Hindi
- Departments of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Guangyan Xiong
- Departments of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Ashok Kumar
- Departments of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
33
|
Sousa-Victor P, Neves J, Muñoz-Cánoves P. Muscle stem cell aging: identifying ways to induce tissue rejuvenation. Mech Ageing Dev 2020; 188:111246. [PMID: 32311419 DOI: 10.1016/j.mad.2020.111246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/26/2020] [Accepted: 04/05/2020] [Indexed: 11/16/2022]
Abstract
Aging is characterized by the functional and regenerative decline of tissues and organs. This regenerative decline is a consequence of the numerical and functional loss of adult stem cells, which are the corner stone of tissue homeostasis and repair. A palpable example of this decline is provided by skeletal muscle, a specialized tissue composed of postmitotic myofibers that contract to generate force. Skeletal muscle stem cells (satellite cells) are long-lived and support muscle regeneration throughout life, but at advanced age they fail for largely undefined reasons. Here, we discuss recent advances in the understanding of how satellite cells integrate diverse intrinsic and extrinsic processes to ensure optimal homeostatic function and how this integration is perturbed during aging, causing regenerative failure. With this increased understanding, it is now feasible to design and test interventions that delay satellite cell aging. We discuss the exciting new therapeutic potential of integrating and combining distinct anti-aging strategies for regenerative medicine.
Collapse
Affiliation(s)
- Pedro Sousa-Victor
- Instituto de Medicina Molecular (iMM), Faculdade de Medicina, Universidade de Lisboa, Lisbon, 1649-028, Portugal.
| | - Joana Neves
- Instituto de Medicina Molecular (iMM), Faculdade de Medicina, Universidade de Lisboa, Lisbon, 1649-028, Portugal.
| | - Pura Muñoz-Cánoves
- Department of Experimental & Health Sciences, University Pompeu Fabra (UPF), CIBERNED, ICREA, 08003, Barcelona, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28019, Madrid, Spain.
| |
Collapse
|
34
|
Pandya VA, Patani R. Decoding the relationship between ageing and amyotrophic lateral sclerosis: a cellular perspective. Brain 2020; 143:1057-1072. [PMID: 31851317 PMCID: PMC7174045 DOI: 10.1093/brain/awz360] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/13/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022] Open
Abstract
With an ageing population comes an inevitable increase in the prevalence of age-associated neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), a relentlessly progressive and universally fatal disease characterized by the degeneration of upper and lower motor neurons within the brain and spinal cord. Indeed, the physiological process of ageing causes a variety of molecular and cellular phenotypes. With dysfunction at the neuromuscular junction implicated as a key pathological mechanism in ALS, and each lower motor unit cell type vulnerable to its own set of age-related phenotypes, the effects of ageing might in fact prove a prerequisite to ALS, rendering the cells susceptible to disease-specific mechanisms. Moreover, we discuss evidence for overlap between age and ALS-associated hallmarks, potentially implicating cell type-specific ageing as a key contributor to this multifactorial and complex disease. With a dearth of disease-modifying therapy currently available for ALS patients and a substantial failure in bench to bedside translation of other potential therapies, the unification of research in ageing and ALS requires high fidelity models to better recapitulate age-related human disease and will ultimately yield more reliable candidate therapeutics for patients, with the aim of enhancing healthspan and life expectancy.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| | - Rickie Patani
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
35
|
Valentine JM, Li ME, Shoelson SE, Zhang N, Reddick RL, Musi N. NFκB Regulates Muscle Development and Mitochondrial Function. J Gerontol A Biol Sci Med Sci 2020; 75:647-653. [PMID: 30423026 PMCID: PMC7328192 DOI: 10.1093/gerona/gly262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Indexed: 11/13/2022] Open
Abstract
Nuclear factor (NF)κB is a transcription factor that controls immune and inflammatory signaling pathways. In skeletal muscle, NFκB has been implicated in the regulation of metabolic processes and tissue mass, yet its affects on mitochondrial function in this tissue are unclear. To investigate the role of NFκB on mitochondrial function and its relationship with muscle mass across the life span, we study a mouse model with muscle-specific NFκB suppression (muscle-specific IκBα super-repressor [MISR] mice). In wild-type mice, there was a natural decline in muscle mass with aging that was accompanied by decreased mitochondrial function and mRNA expression of electron transport chain subunits. NFκB inactivation downregulated expression of PPARGC1A, and upregulated TFEB and PPARGC1B. NFκB inactivation also decreased gastrocnemius (but not soleus) muscle mass in early life (1-6 months old). Lower oxygen consumption rates occurred in gastrocnemius and soleus muscles from young MISR mice, whereas soleus (but not gastrocnemius) muscles from old MISR mice displayed increased oxygen consumption compared to age-matched controls. We conclude that the NFκB pathway plays an important role in muscle development and growth. The extent to which NFκB suppression alters mitochondrial function is age dependent and muscle specific. Finally, mitochondrial function and muscle mass are tightly associated in both genotypes and across the life span.
Collapse
Affiliation(s)
- Joseph M Valentine
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas
| | - Mengyao E Li
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas
- Joslin Diabetes Center, Boston, Massachusetts
| | | | - Ning Zhang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas
| | - Robert L Reddick
- Department of Pathology, University of Texas Health Science Center at San Antonio, Texas
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas
- San Antonio Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
36
|
Stem Cell Aging in Skeletal Muscle Regeneration and Disease. Int J Mol Sci 2020; 21:ijms21051830. [PMID: 32155842 PMCID: PMC7084237 DOI: 10.3390/ijms21051830] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle comprises 30-40% of the weight of a healthy human body and is required for voluntary movements in humans. Mature skeletal muscle is formed by multinuclear cells, which are called myofibers. Formation of myofibers depends on the proliferation, differentiation, and fusion of muscle progenitor cells during development and after injury. Muscle progenitor cells are derived from muscle satellite (stem) cells (MuSCs), which reside on the surface of the myofiber but beneath the basement membrane. MuSCs play a central role in postnatal maintenance, growth, repair, and regeneration of skeletal muscle. In sedentary adult muscle, MuSCs are mitotically quiescent, but are promptly activated in response to muscle injury. Physiological and chronological aging induces MuSC aging, leading to an impaired regenerative capability. Importantly, in pathological situations, repetitive muscle injury induces early impairment of MuSCs due to stem cell aging and leads to early impairment of regeneration ability. In this review, we discuss (1) the role of MuSCs in muscle regeneration, (2) stem cell aging under physiological and pathological conditions, and (3) prospects related to clinical applications of controlling MuSCs.
Collapse
|
37
|
Diaz-Castro J, Moreno-Fernandez J, Chirosa I, Chirosa LJ, Guisado R, Ochoa JJ. Beneficial Effect of Ubiquinol on Hematological and Inflammatory Signaling during Exercise. Nutrients 2020; 12:nu12020424. [PMID: 32041223 PMCID: PMC7071169 DOI: 10.3390/nu12020424] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 01/03/2023] Open
Abstract
Strenuous exercise (any activity that expends six metabolic equivalents per minute or more causing sensations of fatigue and exhaustion to occur, inducing deleterious effects, affecting negatively different cells), induces muscle damage and hematological changes associated with high production of pro-inflammatory mediators related to muscle damage and sports anemia. The objective of this study was to determine whether short-term oral ubiquinol supplementation can prevent accumulation of inflammatory mediators and hematological impairment associated to strenuous exercise. For this purpose, 100 healthy and well-trained firemen were classified in two groups: Ubiquinol (experimental group), and placebo group (control). The protocol was two identical strenuous exercise tests with rest period between tests of 24 h. Blood samples were collected before supplementation (basal value) (T1), after supplementation (T2), after first physical exercise test (T3), after 24 h of rest (T4), and after second physical exercise test (T5). Hematological parameters, pro- and anti-inflammatory cytokines and growth factors were measured. Red blood cells (RBC), hematocrit, hemoglobin, VEGF, NO, EGF, IL-1ra, and IL-10 increased in the ubiquinol group while IL-1, IL-8, and MCP-1 decreased. Ubiquinol supplementation during high intensity exercise could modulate inflammatory signaling, expression of pro-inflammatory, and increasing some anti-inflammatory cytokines. During exercise, RBC, hemoglobin, hematocrit, VEGF, and EGF increased in ubiquinol group, revealing a possible pro-angiogenic effect, improving oxygen supply and exerting a possible protective effect on other physiological alterations.
Collapse
Affiliation(s)
- Javier Diaz-Castro
- Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Biomedical Research Centre, Health-Sciencies Technological Park, Avenida del Conocimiento s/n, Armilla, E-18071 Granada, Spain; (J.M.-F.); (J.J.O.)
- Department of Physiology, University of Granada, E-18071 Granada, Spain
- Correspondence: ; Tel.: +34-958-24-10-00 (ext. 20303)
| | - Jorge Moreno-Fernandez
- Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Biomedical Research Centre, Health-Sciencies Technological Park, Avenida del Conocimiento s/n, Armilla, E-18071 Granada, Spain; (J.M.-F.); (J.J.O.)
- Department of Physiology, University of Granada, E-18071 Granada, Spain
| | - Ignacio Chirosa
- Departament of Physical Education, University of Granada, E-18071 Granada, Spain; (I.C.); (L.J.C.)
| | - Luis Javier Chirosa
- Departament of Physical Education, University of Granada, E-18071 Granada, Spain; (I.C.); (L.J.C.)
| | - Rafael Guisado
- Faculty of Health Sciences, University of Granada, E-18071 Granada, Spain;
| | - Julio J. Ochoa
- Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Biomedical Research Centre, Health-Sciencies Technological Park, Avenida del Conocimiento s/n, Armilla, E-18071 Granada, Spain; (J.M.-F.); (J.J.O.)
- Department of Physiology, University of Granada, E-18071 Granada, Spain
| |
Collapse
|
38
|
Muñoz‐Cánoves P, Neves J, Sousa‐Victor P. Understanding muscle regenerative decline with aging: new approaches to bring back youthfulness to aged stem cells. FEBS J 2020; 287:406-416. [DOI: 10.1111/febs.15182] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Pura Muñoz‐Cánoves
- Department of Experimental and Health Sciences CIBERNED ICREA University Pompeu Fabra (UPF) Barcelona Spain
- Spanish National Cardiovascular Research Center Madrid Spain
| | - Joana Neves
- Instituto de Medicina Molecular (iMM) Faculdade de Medicina Universidade de Lisboa Portugal
| | - Pedro Sousa‐Victor
- Instituto de Medicina Molecular (iMM) Faculdade de Medicina Universidade de Lisboa Portugal
| |
Collapse
|
39
|
Ciuffoli V, Lena AM, Gambacurta A, Melino G, Candi E. Myoblasts rely on TAp63 to control basal mitochondria respiration. Aging (Albany NY) 2019; 10:3558-3573. [PMID: 30487319 PMCID: PMC6286837 DOI: 10.18632/aging.101668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
p53, with its family members p63 and p73, have been shown to promote myoblast differentiation by regulation of the function of the retinoblastoma protein and by direct activation of p21Cip/Waf1 and p57Kip2, promoting cell cycle exit. In previous studies, we have demonstrated that the TAp63γ isoform is the only member of the p53 family that accumulates during in vitro myoblasts differentiation, and that its silencing led to delay in myotube fusion. To better dissect the role of TAp63γ in myoblast physiology, we have generated both sh-p63 and Tet-On inducible TAp63γ clones. Gene array analysis of sh-p63 C2C7 clones showed a significant modulation of genes involved in proliferation and cellular metabolism. Indeed, we found that sh-p63 C2C7 myoblasts present a higher proliferation rate and that, conversely, TAp63γ ectopic expression decreases myoblasts proliferation, indicating that TAp63γ specifically contributes to myoblasts proliferation, independently of p53 and p73. In addition, sh-p63 cells have a defect in mitochondria respiration highlighted by a reduction in spare respiratory capacity and a decrease in complex I, IV protein levels. These results demonstrated that, beside contributing to cell cycle exit, TAp63γ participates to myoblasts metabolism control.
Collapse
Affiliation(s)
- Veronica Ciuffoli
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Biochemistry laboratory, Rome, Italy
| |
Collapse
|
40
|
Smirnov A, Cappello A, Lena AM, Anemona L, Mauriello A, Di Daniele N, Annicchiarico-Petruzzelli M, Melino G, Candi E. ZNF185 is a p53 target gene following DNA damage. Aging (Albany NY) 2019; 10:3308-3326. [PMID: 30446632 PMCID: PMC6286825 DOI: 10.18632/aging.101639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022]
Abstract
The transcription factor p53 is a key player in the tumour suppressive DNA damage response and a growing number of target genes involved in these pathways has been identified. p53 has been shown to be implicated in controlling cell motility and its mutant form enhances metastasis by loss of cell directionality, but the p53 role in this context has not yet being investigated. Here, we report that ZNF185, an actin cytoskeleton-associated protein from LIM-family of Zn-finger proteins, is induced following DNA-damage. ChIP-seq analysis, chromatin crosslinking immune-precipitation experiments and luciferase assays demonstrate that ZNF185 is a bona fide p53 target gene. Upon genotoxic stress, caused by DNA-damaging drug etoposide and UVB irradiation, ZNF185 expression is up-regulated and in etoposide-treated cells, ZNF185 depletion does not affect cell proliferation and apoptosis, but interferes with actin cytoskeleton remodelling and cell polarization. Bioinformatic analysis of different types of epithelial cancers from both TCGA and GTEx databases showed a significant decrease in ZNF185 mRNA level compared to normal tissues. These findings are confirmed by tissue micro-array IHC staining. Our data highlight the involvement of ZNF185 and cytoskeleton changes in p53-mediated cellular response to genotoxic stress and indicate ZNF185 as potential biomarker for epithelial cancer diagnosis.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Angela Cappello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata-IRCCS, Rome 00163, Italy
| |
Collapse
|
41
|
Neves J, Sousa-Victor P. Regulation of inflammation as an anti-aging intervention. FEBS J 2019; 287:43-52. [PMID: 31529582 DOI: 10.1111/febs.15061] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/08/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
Abstract
Aging is accompanied by a decline in physiological integrity and a loss of regenerative capacity in many tissues. The development of interventions that prevent or reverse age-related disease requires a better understanding of the interplay of cell intrinsic, inter-cellular communication and systemic deregulations that underlie the aging process. Immune dysfunction and changes in inflammatory pathways are transversal contributors to the aging process and are essential propagators of tissue deterioration. Here, we propose and discuss the rejuvenation potential of interventions that target chronic inflammation and how modulation of tissue repair capacity could be an important mediator of such anti-aging strategies. We highlight how current knowledge on the systemic nature of inflammatory dysregulation in old organisms, together with the development of new animal models that allow for the isolation of the inflammatory component of aging, could provide new targets for interventions in aging based on the modulation of inflammatory pathways.
Collapse
Affiliation(s)
- Joana Neves
- Faculdade de Medicina, Instituto de Medicina Molecular (iMM), Universidade de Lisboa, Portugal
| | - Pedro Sousa-Victor
- Faculdade de Medicina, Instituto de Medicina Molecular (iMM), Universidade de Lisboa, Portugal
| |
Collapse
|
42
|
Budai Z, Balogh L, Sarang Z. Altered Gene Expression of Muscle Satellite Cells Contributes to Agerelated Sarcopenia in Mice. Curr Aging Sci 2019; 11:165-172. [PMID: 30251615 PMCID: PMC6388427 DOI: 10.2174/1874609811666180925104241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/21/2018] [Accepted: 07/21/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND During aging, muscle tissue undergoes profound changes which lead to a decline in its functional and regenerative capacity. We utilized global gene expression analysis and gene set enrichment analysis to characterize gene expression changes in aging muscle satellite cells. METHOD Gene expression data; obtained from Affymetrix Mouse Genome 430 2.0 Array, for 14 mouse muscle satellite cell samples (5 young, 4 middle-aged, and 5 aged), were retrieved from public Gene Expression Omnibus repository. List of differentially expressed genes was generated based on 0.05 multiple-testing-adjusted p-value and 2-fold FC cut-off values. Functional profiling of genes was carried out using PANTHER Classification System. RESULTS We have found several differentially expressed genes in satellite cells derived from aged mice compared to young ones. The gene expression changes increased progressively with time, and the majority of the differentially expressed genes were upregulated during aging. While the downregulated genes could not be correlated with specific biological processes the upregulated ones could be associated with muscle differentiation-, inflammation- or fibrosis-related processes. The latter two processes encompass the senescence-associated secretory phenotype for satellite cells which alters the tissue microenvironment and contributes to inflammation and fibrosis observed in aging muscle. CONCLUSION Our analysis reveals that by altering gene expression pattern and expressing inflammatory mediators and extracellular matrix components, these cells can directly contribute to muscle wasting in aged mice.
Collapse
Affiliation(s)
- Zsofia Budai
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Laszlo Balogh
- Institute of Sport Sciences, University of Debrecen, Debrecen, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
43
|
Wang MJ, Chen J, Chen F, Liu Q, Sun Y, Yan C, Yang T, Bao Y, Hu YP. Rejuvenating Strategies of Tissue-specific Stem Cells for Healthy Aging. Aging Dis 2019; 10:871-882. [PMID: 31440391 PMCID: PMC6675530 DOI: 10.14336/ad.2018.1119] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
Although aging is a physiological process, it has raised interest in the science of aging and rejuvenation because of the increasing burden on the rapidly aging global population. With advanced age, there is a decline in homeostatic maintenance and regenerative responsiveness to the injury of various tissues, thereby contributing to the incidence of age-related diseases. The primary cause of the functional declines that occur along with aging is considered to be the exhaustion of stem cell functions in their corresponding tissues. Age-related changes in the systemic environment, the niche, and stem cells contribute to this loss. Thus, the reversal of stem cell aging at the cellular level might lead to the rejuvenation of the animal at an organismic level and the prevention of aging, which would be critical for developing new therapies for age-related dysfunction and diseases. Here, we will explore the effects of aging on stem cells in different tissues. The focus of this discussion is on pro-youth interventions that target intrinsic stem cell properties, environmental niche component, systemic factors, and senescent cellular clearance, which are promising for developing strategies related to the reversal of aged stem cell function and optimizing tissue repair processes.
Collapse
Affiliation(s)
- Min-Jun Wang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Jiajia Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Fei Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Qinggui Liu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yu Sun
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chen Yan
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Tao Yang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yiwen Bao
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China.,2Department of Diagnostic radiology, University of Hong Kong, Hong Kong 999077, China
| | - Yi-Ping Hu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
44
|
Zhu P, Zhang C, Gao Y, Wu F, Zhou Y, Wu WS. The transcription factor Slug represses p16 Ink4a and regulates murine muscle stem cell aging. Nat Commun 2019; 10:2568. [PMID: 31189923 PMCID: PMC6561969 DOI: 10.1038/s41467-019-10479-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/14/2019] [Indexed: 01/21/2023] Open
Abstract
Activation of the p16Ink4a-associated senescence pathway during aging breaks muscle homeostasis and causes degenerative muscle disease by irreversibly dampening satellite cell (SC) self-renewal capacity. Here, we report that the zinc-finger transcription factor Slug is highly expressed in quiescent SCs of mice and functions as a direct transcriptional repressor of p16Ink4a. Loss of Slug promotes derepression of p16Ink4a in SCs and accelerates the entry of SCs into a fully senescent state upon damage-induced stress. p16Ink4a depletion partially rescues defects in Slug-deficient SCs. Furthermore, reduced Slug expression is accompanied by p16Ink4a accumulation in aged SCs. Slug overexpression ameliorates aged muscle regeneration by enhancing SC self-renewal through active repression of p16Ink4a transcription. Our results identify a cell-autonomous mechanism underlying functional defects of SCs at advanced age. As p16Ink4a dysregulation is the chief cause for regenerative defects of human geriatric SCs, these findings highlight Slug as a potential therapeutic target for aging-associated degenerative muscle disease. Muscle regeneration depends on self-renewal of muscle stem cells but how this is regulated on aging is unclear. Here, the authors identify Slug as regulating p16Ink4a in quiescent muscle stem cells, and when Slug expression reduces in aged stem cells, p16Ink4a accumulates, causing regenerative defects.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Chunping Zhang
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yongxing Gao
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Furen Wu
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yalu Zhou
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Wen-Shu Wu
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
45
|
Metabolic and Molecular Basis of Sarcopenia: Implications in the Management of Urothelial Carcinoma. Int J Mol Sci 2019; 20:ijms20030760. [PMID: 30754663 PMCID: PMC6387186 DOI: 10.3390/ijms20030760] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/05/2019] [Accepted: 02/09/2019] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia, which represents the degenerative and systemic loss of skeletal muscle mass, is a multifactorial syndrome caused by various clinical conditions. Sarcopenia reflects not only frailty and poor general health status, but also the possible presence of advanced or progressive cancer or cancer cachexia. Therefore, sarcopenia affects the management of cancer-bearing patients, including those with urothelial carcinoma. Recently, growing evidence has shown that sarcopenia is significantly associated with higher rates of treatment-related complications and worse prognosis in patients with urothelial carcinoma, including muscle-invasive bladder cancer, upper tract urothelial carcinoma, and advanced urothelial carcinoma. Moreover, several studies reported that a post-therapeutic increase in skeletal muscle mass predicts favorable prognosis in urothelial carcinoma patients. To further explore the role of sarcopenia in the management of urothelial carcinoma patients, comprehensive understanding of its pathophysiology is vital. In this article, we reviewed the metabolic and molecular basis of cancer cachexia and sarcopenia. From this viewpoint, we discussed the possible mechanism of changes in skeletal muscle mass during the course of treatment.
Collapse
|
46
|
Momtaz S, Baeeri M, Rahimifard M, Haghi‐Aminjan H, Hassani S, Abdollahi M. Manipulation of molecular pathways and senescence hallmarks by natural compounds in fibroblast cells. J Cell Biochem 2018; 120:6209-6222. [DOI: 10.1002/jcb.27909] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences Tehran Iran
| | - Maryam Baeeri
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
| | - Mahban Rahimifard
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
| | - Hamed Haghi‐Aminjan
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
- Department of Toxicology and Pharmacology Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
| | - Shokoufeh Hassani
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
| | - Mohammad Abdollahi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR Karaj Iran
- Department of Toxicology and Pharmacology Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
47
|
Obese subcutaneous adipose tissue impairs human myogenesis, particularly in old skeletal muscle, via resistin-mediated activation of NFκB. Sci Rep 2018; 8:15360. [PMID: 30337633 PMCID: PMC6193975 DOI: 10.1038/s41598-018-33840-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/05/2018] [Indexed: 02/02/2023] Open
Abstract
Adiposity and adipokines are implicated in the loss of skeletal muscle mass with age and in several chronic disease states. The aim of this study was to determine the effects of human obese and lean subcutaneous adipose tissue secretome on myogenesis and metabolism in skeletal muscle cells derived from both young (18-30 yr) and elderly (>65 yr) individuals. Obese subcutaneous adipose tissue secretome impaired the myogenesis of old myoblasts but not young myoblasts. Resistin was prolifically secreted by obese subcutaneous adipose tissue and impaired myotube thickness and nuclear fusion by activation of the classical NFκB pathway. Depletion of resistin from obese adipose tissue secretome restored myogenesis. Inhibition of the classical NFκB pathway protected myoblasts from the detrimental effect of resistin on myogenesis. Resistin also promoted intramyocellular lipid accumulation in myotubes and altered myotube metabolism by enhancing fatty acid oxidation and increasing myotube respiration and ATP production. In conclusion, resistin derived from human obese subcutaneous adipose tissue impairs myogenesis of human skeletal muscle, particularly older muscle, and alters muscle metabolism in developing myotubes. These findings may have important implications for the maintenance of muscle mass in older people with chronic inflammatory conditions, or older people who are obese or overweight.
Collapse
|
48
|
Todhunter ME, Sayaman RW, Miyano M, LaBarge MA. Tissue aging: the integration of collective and variant responses of cells to entropic forces over time. Curr Opin Cell Biol 2018; 54:121-129. [PMID: 29908481 PMCID: PMC6258070 DOI: 10.1016/j.ceb.2018.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/13/2018] [Accepted: 05/30/2018] [Indexed: 01/08/2023]
Abstract
Aging is driven by unavoidable entropic forces, physicochemical in nature, that damage the raw materials that constitute biological systems. Single cells experience and respond to stochastic physicochemical insults that occur either to the cells themselves or to their microenvironment, in a dynamic and reciprocal manner, leading to increased age-related cell-to-cell variation. We will discuss the biological mechanisms that integrate cell-to-cell variation across tissues resulting in stereotypical phenotypes of age.
Collapse
Affiliation(s)
- Michael E Todhunter
- Department of Populations Sciences, and Center for Cancer and Aging, Beckman Research Institute at City of Hope, 1500 E Duarte Rd, Duarte, CA 91011 USA
| | - Rosalyn W Sayaman
- Department of Populations Sciences, and Center for Cancer and Aging, Beckman Research Institute at City of Hope, 1500 E Duarte Rd, Duarte, CA 91011 USA
| | - Masaru Miyano
- Department of Populations Sciences, and Center for Cancer and Aging, Beckman Research Institute at City of Hope, 1500 E Duarte Rd, Duarte, CA 91011 USA
| | - Mark A LaBarge
- Department of Populations Sciences, and Center for Cancer and Aging, Beckman Research Institute at City of Hope, 1500 E Duarte Rd, Duarte, CA 91011 USA.
| |
Collapse
|
49
|
|
50
|
Dinulovic I, Furrer R, Handschin C. Plasticity of the Muscle Stem Cell Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:141-169. [PMID: 29204832 DOI: 10.1007/978-3-319-69194-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Satellite cells (SCs) are adult muscle stem cells capable of repairing damaged and creating new muscle tissue throughout life. Their functionality is tightly controlled by a microenvironment composed of a wide variety of factors, such as numerous secreted molecules and different cell types, including blood vessels, oxygen, hormones, motor neurons, immune cells, cytokines, fibroblasts, growth factors, myofibers, myofiber metabolism, the extracellular matrix and tissue stiffness. This complex niche controls SC biology-quiescence, activation, proliferation, differentiation or renewal and return to quiescence. In this review, we attempt to give a brief overview of the most important players in the niche and their mutual interaction with SCs. We address the importance of the niche to SC behavior under physiological and pathological conditions, and finally survey the significance of an artificial niche both for basic and translational research purposes.
Collapse
|