1
|
Traoré M, Noviello C, Vergnol A, Gentil C, Halliez M, Saillard L, Gelin M, Forand A, Lemaitre M, Guesmia Z, Cadot B, Caldas de Almeida Araujo E, Marty B, Mougenot N, Messéant J, Strochlic L, Sadoine J, Slimani L, Jolly A, De la Grange P, Hogrel JY, Pietri-Rouxel F, Falcone S. GDF5 as a rejuvenating treatment for age-related neuromuscular failure. Brain 2024; 147:3834-3848. [PMID: 38584513 DOI: 10.1093/brain/awae107] [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: 11/22/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/09/2024] Open
Abstract
Sarcopenia involves a progressive loss of skeletal muscle force, quality and mass during ageing, which results in increased inability and death; however, no cure has been established thus far. Growth differentiation factor 5 (GDF5) has been described to modulate muscle mass maintenance in various contexts. For our proof of concept, we overexpressed GDF5 by AAV vector injection in tibialis anterior muscle of adult aged (20 months) mice and performed molecular and functional analysis of skeletal muscle. We analysed human vastus lateralis muscle biopsies from adult young (21-42 years) and aged (77-80 years) donors, quantifying the molecular markers modified by GDF5 overexpression in mouse muscle. We validated the major effects of GDF5 overexpression using human immortalized myotubes and Schwann cells. We established a preclinical study by treating chronically (for 4 months) aged mice using recombinant GDF5 protein (rGDF5) in systemic administration and evaluated the long-term effect of this treatment on muscle mass and function. Here, we demonstrated that GDF5 overexpression in the old tibialis anterior muscle promoted an increase of 16.5% of muscle weight (P = 0.0471) associated with a higher percentage of 5000-6000 µm2 large fibres (P = 0.0211), without the induction of muscle regeneration. Muscle mass gain was associated with an amelioration of 26.8% of rate of force generation (P = 0.0330) and better neuromuscular connectivity (P = 0.0098). Moreover, GDF5 overexpression preserved neuromuscular junction morphology (38.5% of nerve terminal area increase, P < 0.0001) and stimulated the expression of reinnervation-related genes, in particular markers of Schwann cells (fold-change 3.19 for S100b gene expression, P = 0.0101). To characterize the molecular events induced by GDF5 overexpression during ageing, we performed a genome-wide transcriptomic analysis of treated muscles and showed that this factor leads to a 'rejuvenating' transcriptomic signature in aged mice, as 42% of the transcripts dysregulated by ageing reverted to youthful expression levels upon GDF5 overexpression (P < 0.05). Towards a preclinical approach, we performed a long-term systemic treatment using rGDF5 and showed its effectiveness in counteracting age-related muscle wasting, improving muscle function (17.8% of absolute maximal force increase, P = 0.0079), ensuring neuromuscular connectivity and preventing neuromuscular junction degeneration (7.96% of AchR area increase, P = 0.0125). In addition, in human muscle biopsies, we found the same age-related alterations than those observed in mice and improved by GDF5 and reproduced its major effects on human cells, suggesting this treatment as efficient in humans. Overall, these data provide a foundation to examine the curative potential of GDF5 drug in clinical trials for sarcopenia and, eventually, other neuromuscular diseases.
Collapse
Affiliation(s)
- Massiré Traoré
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Chiara Noviello
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Amélie Vergnol
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Christel Gentil
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marius Halliez
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Lucile Saillard
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Maxime Gelin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Anne Forand
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
- Inovarion, F-75005 Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Bruno Cadot
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | | | - Benjamin Marty
- Institut de Myologie, CEA, Laboratoire d'imagerie et de spectroscopie par RMN, F-75013 Paris, France
| | - Nathalie Mougenot
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Julien Messéant
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Laure Strochlic
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Jeremy Sadoine
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Lofti Slimani
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Ariane Jolly
- GenoSplice, Paris Biotech Santé, F-75014 Paris, France
| | | | - Jean-Yves Hogrel
- Institut de Myologie, Laboratoire de physiologie et d'évaluation neuromusculaire, F-75013 Paris, France
| | - France Pietri-Rouxel
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| |
Collapse
|
2
|
Hung YL, Sato A, Takino Y, Ishigami A, Machida S. Resistance training suppresses accumulation of senescent fibro-adipogenic progenitors and senescence-associated secretory phenotype in aging rat skeletal muscle. GeroScience 2024:10.1007/s11357-024-01338-2. [PMID: 39298108 DOI: 10.1007/s11357-024-01338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Accumulation of senescent cells in tissues contributes to multiple aging-related pathologies. Senescent fibro-adipogenic progenitors (FAPs) contribute to aging-related muscle atrophy. Resistance training can help to maintain skeletal muscle mass, improve mobility, and reduce certain health risks commonly associated with aging. We investigated, using rat model, the impact of resistance training on FAPs in aging skeletal muscle, which remains unclear. Twenty-two-month-old female rats were divided into sedentary and training groups. The training group rodents were trained to climb a ladder while bearing a load for 20 training sessions over 2 months, after which, the flexor hallucis longus muscles were collected and analyzed. Senescent cells were identified using a senescence-associated β-galactosidase stain and p21 immunohistochemistry (IHC), and FAPs were identified using platelet-derived growth factor receptor alpha IHC. The results indicate that resistance training in rats prevented aging-associated skeletal muscle atrophy and suppressed M2 polarization of macrophages. The number of senescent cells was significantly reduced in the 24-month-old training group, with most of them being FAPs. Conversely, the number of senescent FAPs increased significantly in the 24-month-old sedentary group compared with that in the 18-month-old sedentary group. The number of senescent FAPs in the 24-month-old training group decreased significantly. Resistance training also suppressed the senescence-associated secretory phenotype (SASP). The killer T cell-specific marker, CD8α, was elevated in the skeletal muscles of the aging rats following resistance training, indicating upregulation of recognition and elimination of senescent cells. Overall, resistance training suppressed the accumulation of senescent FAPs and acquisition of SASP in aging skeletal muscles.
Collapse
Affiliation(s)
- Yung-Li Hung
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-Ku, Tokyo, 102-0083, Japan
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
- Institute of Health and Sports Science & Medicine, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| |
Collapse
|
3
|
Dunham CL, Frank JA. Ultrasound Pressure-Dependent Cytokine and Immune Cell Response Lost in Aged Muscle. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:494-501. [PMID: 38218683 PMCID: PMC10922560 DOI: 10.1016/j.ultrasmedbio.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 12/05/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVE Therapeutic ultrasound remains a highly discussed topic in physical therapy due to uncertainty between treatment regimens and biological benefits. Its impact on aged populations, who are vulnerable to insufficient healing after muscle injury because of sarcopenia, is understudied. Despite the coupling between muscle inflammation and regeneration, research on the immune response after therapeutic ultrasound is limited. The objective of this study was to evaluate structure, inflammatory cytokine signaling and immune cell infiltration after therapeutic ultrasound in young and aging murine muscle. METHODS Young (6-week-old) and Adult (52-week-old) male and female mouse non-injured gastrocnemii were treated with either low-intensity pulsed ultrasound at 2 W/cm2 (∼0.243 MPa) or high-intensity pulsed focused ultrasound at 554 W/cm2 (∼5.96 MPa). Cytokine expression was evaluated at 1, 8 and 24 hours, cell infiltration was measured via flow cytometry at 1 and 24 hours and immunofluorescence assessed muscle fiber area, fibrosis and satellite cells at 24 hours after sonication. RESULTS Low-intensity pulsed ultrasound induced an early, transient inflammatory response where interleukin (IL)-15 and macrophages (M2 > M1) were increased 1 hour post-sonication. High-intensity pulsed focused ultrasound caused a late, extended immune response where monocyte chemoattractant protein 1 (MCP-1), neutrophils, monocytes and macrophages (M1 > M2) were increased 24 hours post-sonication. Notably, these changes manifested solely in Young gastrocnemius. The Adult gastrocnemius exhibited decreased cytokine expression (IL-1α, IL-6, IL-15, macrophage colony-stimulating factor [M-CSF]) and no alteration in immune cell recruitment post-sonication. There was no damage to muscle structure. CONCLUSION Therapeutic ultrasound induced a pressure-dependent inflammatory response that can augment or mitigate intrinsic muscle cytokine signaling and cell recruitment in adolescent or aged muscle, respectively.
Collapse
Affiliation(s)
- Chelsey L Dunham
- Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| | - Joseph A Frank
- Clinical Center, National Institutes of Health, Bethesda, MD, USA; National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
A cross-talk between sestrins, chronic inflammation and cellular senescence governs the development of age-associated sarcopenia and obesity. Ageing Res Rev 2023; 86:101852. [PMID: 36642190 DOI: 10.1016/j.arr.2023.101852] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The rapid increase in both the lifespan and proportion of older adults is accompanied by the unprecedented rise in age-associated chronic diseases, including sarcopenia and obesity. Aging is also manifested by increased susceptibility to multiple endogenous and exogenous stresses enabling such chronic conditions to develop. Among the main physiological regulators of cellular adaption to various stress stimuli, such as DNA damage, hypoxia, and oxidative stress, are sestrins (Sesns), a family of three evolutionarily conserved proteins, Sesn1, 2, and 3. Age-associated sarcopenia and obesity are characterized by two key processes: (i) accumulation of senescent cells in the skeletal muscle and adipose tissue and (ii) creation of a systemic, chronic, low-grade inflammation (SCLGI). Presumably, failed SCLGI resolution governs the development of these chronic conditions. Noteworthy, Sesns activate senolytics, which are agents that selectively eliminate senescent cells, as well as specialized pro-resolving mediators, which are factors that physiologically provide inflammation resolution. Sesns reveal clear beneficial effects in pre-clinical models of sarcopenia and obesity. Based on these observations, we propose a novel treatment strategy for age-associated sarcopenia and obesity, complementary to the conventional therapeutic modalities: Sesn activation, SCLGI resolution, and senescent cell elimination.
Collapse
|
5
|
Nakanishi N, Ono Y, Miyazaki Y, Moriyama N, Fujioka K, Yamashita K, Inoue S, Kotani J. Sepsis causes neutrophil infiltration in muscle leading to muscle atrophy and weakness in mice. Front Immunol 2022; 13:950646. [PMID: 36389802 PMCID: PMC9659852 DOI: 10.3389/fimmu.2022.950646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/19/2022] [Indexed: 12/03/2022] Open
Abstract
Background Sepsis-induced muscle atrophy leads to prolonged physical dysfunction. Although the interaction of muscle atrophy and macrophage has been reported in sepsis, the role of neutrophils in muscle atrophy has not been thoroughly investigated. This study sought to investigate the long-term changes in muscle-localized neutrophils after sepsis induction and their possible role in sepsis. Methods Sepsis was induced in seven-week-old male C57BL/6J mice 8-12 (cecal slurry [CS] model) via intraperitoneal injection of 1 mg/g cecal slurry. The percentage change in body weight and grip strength was evaluated. The tibialis anterior muscles were dissected for microscopic examination of the cross-sectional area of myofibers or Fluorescence-activated cell sorting (FACS) analysis of immune cells. These changes were evaluated in the following conditions: (1) Longitudinal change until day 61, (2) CS concentration-dependent change on day 14 at the low (0.3 mg/g), middle (1.0 mg/g), and high (2.0 mg/g) concentrations, and (3) CS mice on day 14 treated with an anti-Ly6G antibody that depletes neutrophils. Results Body weight and grip strength were significantly lower in the CS model until day 61 (body weight: 123.1% ± 1.8% vs. 130.3% ± 2.5%, p = 0.04; grip strength: 104.5% ± 3.8% vs. 119.3% ± 5.3%, p = 0.04). Likewise, cross-sectional muscle area gradually decreased until day 61 from the CS induction (895.6 [606.0–1304.9] μm2 vs. 718.8 [536.2–937.0] μm2, p < 0.01). The number of muscle-localized neutrophils increased from 2.3 ± 0.6 cell/mg on day 0 to 22.2 ± 13.0 cell/mg on day 14, and decreased thereafter. In terms of CS concentration–dependent change, cross-sectional area was smaller (484.4 ± 221.2 vs. 825.8 ± 436.2 μm2 [p < 0.001]) and grip strength was lower (71.4% ± 12.8% vs. 116.3% ± 7.4%, p = 0.01) in the CS High group compared with the control, with increased neutrophils (p = 0.03). Ly6G-depleted mice demonstrated significant increase of muscle cross-sectional area and grip strength compared with control mice (p < 0.01). Conclusions Sepsis causes infiltration of neutrophils in muscles, leading to muscle atrophy and weakness. Depletion of neutrophils in muscle reverses sepsis-induced muscle atrophy and weakness. These results suggest that neutrophils may play a critical role in sepsis-induced muscle atrophy and weakness.
Collapse
Affiliation(s)
- Nobuto Nakanishi
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuko Ono
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusuke Miyazaki
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Moriyama
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigeaki Inoue
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
- *Correspondence: Shigeaki Inoue,
| | - Joji Kotani
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
6
|
Englund DA, Zhang X, Aversa Z, LeBrasseur NK. Skeletal muscle aging, cellular senescence, and senotherapeutics: Current knowledge and future directions. Mech Ageing Dev 2021; 200:111595. [PMID: 34742751 PMCID: PMC8627455 DOI: 10.1016/j.mad.2021.111595] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022]
Abstract
Cellular senescence is a state of cell cycle arrest induced by several forms of metabolic stress. Senescent cells accumulate with advancing age and have a distinctive phenotype, characterized by profound chromatin alterations and a robust senescence-associated secretory phenotype (SASP) that exerts negative effects on tissue health, both locally and systemically. In preclinical models, pharmacological agents that eliminate senescent cells (senotherapeutics) restore health and youthful properties in multiple tissues. To date, however, very little is understood about the vulnerability of terminally-differentiated skeletal muscle fibers and the resident mononuclear cells that populate the interstitial microenvironment of skeletal muscle to senescence, and their contribution to the onset and progression of skeletal muscle loss and dysfunction with aging. Scientific advances in these areas have the potential to highlight new therapeutic approaches to optimize late-life muscle health. To this end, this review highlights the current evidence and the key questions that need to be addressed to advance the field's understanding of cellular senescence as a mediator of skeletal muscle aging and the potential for emerging senescent cell-targeting therapies to counter age-related deficits in muscle mass, strength, and function. This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.
Collapse
Affiliation(s)
- Davis A Englund
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
7
|
Costache AD, Costache II, Miftode RȘ, Stafie CS, Leon-Constantin MM, Roca M, Drugescu A, Popa DM, Mitu O, Mitu I, Miftode LI, Iliescu D, Honceriu C, Mitu F. Beyond the Finish Line: The Impact and Dynamics of Biomarkers in Physical Exercise-A Narrative Review. J Clin Med 2021; 10:jcm10214978. [PMID: 34768497 PMCID: PMC8584497 DOI: 10.3390/jcm10214978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
The research of biomarkers continues to emerge as a developing academic field which is attracting substantial interest. The study of biomarkers proves to be useful in developing and implementing new screening methods for a wide variety of diseases including in the sports area, whether for leisure activities or professional sports. Novel research has brought into question the immune system and the limitations it may impose on sports practicing. As the well-being of athletes is a priority, the state of their immune function offers valuable information regarding their health status and their ability to continue training. The assessment of various biomarkers may contribute to a more accurate risk stratification and subsequent prevention of some invalidating or even fatal pathologies such as the sudden cardiac death. Therefore, we have reviewed several studies that included sports-related pathology or specific morphofunctional alterations for which some immune biomarkers may represent an expression of the underlying mechanism. These include the defensins, immunoglobulin A (IgA), interleukin-6 (IL-6), the tumoral necrosis factor α (TNF-α), and the white blood cells (WBC) count. Similarly, also of significant interest are various endocrine biomarkers, such as cortisol and testosterone, as well as anabolic or catabolic markers, respectively. Literature data highlight that these values are greatly influenced not only by the duration, but also by the intensity of the physical exercise; moderate training sessions actually enhance the immune function of the body, while a significant increase in both duration and intensity of sports activity acts as a deleterious factor. Therefore, in this paper we aim to highlight the importance of biomarkers’ evaluation in connection with sports activities and a subsequent more adequate approach towards personalized training regimens.
Collapse
Affiliation(s)
- Alexandru-Dan Costache
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
| | - Irina-Iuliana Costache
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.-I.C.); (D.-M.P.); (O.M.); (D.I.)
| | - Radu-Ștefan Miftode
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
- Correspondence:
| | - Celina-Silvia Stafie
- Department of Preventive Medicine and Interdisciplinarity, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Maria-Magdalena Leon-Constantin
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
| | - Mihai Roca
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
| | - Andrei Drugescu
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
| | - Delia-Melania Popa
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.-I.C.); (D.-M.P.); (O.M.); (D.I.)
| | - Ovidiu Mitu
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.-I.C.); (D.-M.P.); (O.M.); (D.I.)
| | - Ivona Mitu
- Department of Morpho-Functional Sciences II, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Larisa-Ionela Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Dan Iliescu
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (I.-I.C.); (D.-M.P.); (O.M.); (D.I.)
| | - Cezar Honceriu
- Faculty of Physical Education and Sports, “Alexandru Ioan Cuza” University, 700115 Iasi, Romania;
| | - Florin Mitu
- Department of Cardiovascular Rehabilitation, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-D.C.); (M.-M.L.-C.); (M.R.); (A.D.); (F.M.)
| |
Collapse
|
8
|
Markworth JF, Brown LA, Lim E, Castor‐Macias JA, Larouche J, Macpherson PCD, Davis C, Aguilar CA, Maddipati KR, Brooks SV. Metabolipidomic profiling reveals an age-related deficiency of skeletal muscle pro-resolving mediators that contributes to maladaptive tissue remodeling. Aging Cell 2021; 20:e13393. [PMID: 34075679 PMCID: PMC8208786 DOI: 10.1111/acel.13393] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/07/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022] Open
Abstract
Specialized pro-resolving mediators actively limit inflammation and support tissue regeneration, but their role in age-related muscle dysfunction has not been explored. We profiled the mediator lipidome of aging muscle via liquid chromatography-tandem mass spectrometry and tested whether treatment with the pro-resolving mediator resolvin D1 (RvD1) could rejuvenate the regenerative ability of aged muscle. Aged mice displayed chronic muscle inflammation and this was associated with a basal deficiency of pro-resolving mediators 8-oxo-RvD1, resolvin E3, and maresin 1, as well as many anti-inflammatory cytochrome P450-derived lipid epoxides. Following muscle injury, young and aged mice produced similar amounts of most pro-inflammatory eicosanoid metabolites of cyclooxygenase (e.g., prostaglandin E2 ) and 12-lipoxygenase (e.g., 12-hydroxy-eicosatetraenoic acid), but aged mice produced fewer markers of pro-resolving mediators including the lipoxins (15-hydroxy-eicosatetraenoic acid), D-resolvins/protectins (17-hydroxy-docosahexaenoic acid), E-resolvins (18-hydroxy-eicosapentaenoic acid), and maresins (14-hydroxy-docosahexaenoic acid). Similar absences of downstream pro-resolving mediators including lipoxin A4 , resolvin D6, protectin D1/DX, and maresin 1 in aged muscle were associated with greater inflammation, impaired myofiber regeneration, and delayed recovery of strength. Daily intraperitoneal injection of RvD1 had minimal impact on intramuscular leukocyte infiltration and myofiber regeneration but suppressed inflammatory cytokine expression, limited fibrosis, and improved recovery of muscle function. We conclude that aging results in deficient local biosynthesis of specialized pro-resolving mediators in muscle and that immunoresolvents may be attractive novel therapeutics for the treatment of muscular injuries and associated pain in the elderly, due to positive effects on recovery of muscle function without the negative side effects on tissue regeneration of non-steroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
- James F. Markworth
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
| | - Lemuel A. Brown
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
| | - Eunice Lim
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
| | | | - Jacqueline Larouche
- Department of Biomedical Engineering University of Michigan Ann Arbor MI USA
| | - Peter C. D. Macpherson
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
| | - Carol Davis
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
| | - Carlos A. Aguilar
- Department of Biomedical Engineering University of Michigan Ann Arbor MI USA
| | - Krishna Rao Maddipati
- Department of Pathology Lipidomics Core Facility Wayne State University Detroit MI USA
| | - Susan V. Brooks
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
- Department of Biomedical Engineering University of Michigan Ann Arbor MI USA
| |
Collapse
|
9
|
Kawanishi N, Machida S. Alterations of macrophage and neutrophil content in skeletal muscle of aged versus young mice. Muscle Nerve 2021; 63:600-607. [PMID: 33386611 PMCID: PMC8048435 DOI: 10.1002/mus.27158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/19/2020] [Accepted: 12/24/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Skeletal muscle inflammation and oxidative stress are associated with aging-related loss of muscle mass and may be attributable to alterations in the number and types of leukocytes in skeletal muscle. Here, we tested the hypothesis that aging changes the number and composition of leukocyte subsets in skeletal muscle tissue. METHODS Skeletal muscle was sampled from 4-mo-old (young) and 27-mo-old (old) C57BL/6J mice. Mononuclear cells of the gastrocnemius muscle were isolated, and flow cytometry was used to characterize the number and types of immune cells. RESULTS The number of neutrophils and Ly-6C+ inflammatory macrophages in the skeletal muscle was significantly higher in old mice than in young mice. Inflammation and oxidative stress (measured using the markers phosphorylated JNK and nitrotyrosine) were also higher in the skeletal muscle of old mice than in that of young mice. CONCLUSIONS Increasing age promotes skeletal muscle inflammation and oxidative stress, as well as infiltration of inflammatory macrophages and neutrophils.
Collapse
Affiliation(s)
- Noriaki Kawanishi
- Faculty of Advanced EngineeringChiba Institute of TechnologyNarashinoJapan
- Graduate School of Health and Sports ScienceJuntendo UniversityInzaiJapan
- Institute of Health & Sports Science and MedicineJuntendo UniversityInzaiJapan
| | - Shuichi Machida
- Graduate School of Health and Sports ScienceJuntendo UniversityInzaiJapan
- Institute of Health & Sports Science and MedicineJuntendo UniversityInzaiJapan
| |
Collapse
|