1
|
Alcalde-Estévez E, Moreno-Piedra A, Asenjo-Bueno A, Martos-Elvira M, de la Serna-Soto M, Ruiz-Ortega M, Olmos G, López-Ongil S, Ruiz-Torres MP. Aging-related hyperphosphatemia triggers the release of TNF-α from macrophages, promoting indicators of sarcopenia through the reduction of IL-15 expression in skeletal muscle. Life Sci 2025; 368:123507. [PMID: 40010633 DOI: 10.1016/j.lfs.2025.123507] [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: 09/13/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
AIMS The association between aging-related hyperphosphatemia and sarcopenia has been documented, and evidence suggests that inflammaging is involved in the manifestation of sarcopenia. The present study investigates whether hyperphosphatemia triggers inflammation, thereby inducing the appearance of sarcopenia along with the cytokines involved in these processes. MATERIALS AND METHODS RAW 264.7 macrophages were incubated with β-glycerophosphate (BGP), as a phosphate donor, at different time intervals, to assess the production of proinflammatory markers. Conditioned medium from macrophages was collected and added to cultured C2C12 myoblasts to analyse whether proinflammatory molecules, released by macrophages, modified myogenic differentiation, cell senescence or myokine IL-15 expression. A neutralising antibody anti-TNF-α and recombinant IL-15 were added to evaluate the role of these cytokines in the observed effects. Additionally, TNF-α, IL-15, serum phosphate, and sarcopenia signs were evaluated in 5-month-old mice, 24-month-old mice and 24-month-old mice fed with a hypophosphatemic diet. KEY FINDINGS BGP increased TNF-α expression in macrophages through NFkB activation. Conditioned medium from BGP-treated macrophages impaired myogenic differentiation in differentiating myoblasts and promoted cellular senescence and reduced IL-15 expression in undifferentiated myoblasts. These effects were mediated by TNF-α. Old mice displayed reduced expression of muscle IL-15 and elevated circulating TNF-α, along with increased serum phosphate levels, which correlated with the appearance of sarcopenia indicators. The hypophosphatemic diet prevented these changes in old mice. SIGNIFICANCE Hyperphosphatemia induces TNF-α production in macrophages, which contributes to the reduced expression of muscular IL-15. This mechanism may play a role in inducing sarcopenia in elderly mice.
Collapse
Affiliation(s)
- Elena Alcalde-Estévez
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Ariadna Moreno-Piedra
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Ramón y Cajal Health Research Institute (IRYCIS), Madrid 28034, Spain
| | - Ana Asenjo-Bueno
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - María Martos-Elvira
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Mariano de la Serna-Soto
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology, Institute of Medical Research of the Jiménez Díaz Foundation, Autonomous University of Madrid, Madrid, Spain
| | - Gemma Olmos
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain
| | - Susana López-Ongil
- Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain; Foundation for Biomedical Research of the Príncipe de Asturias University Hospital, Alcalá de Henares, Madrid 28805, Spain
| | - María P Ruiz-Torres
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain
| |
Collapse
|
2
|
Han MM, Liu Q, Yang L, Li J, Wang QY, Zhang YM, Guo JQ, Zeng LT, Qi RM, Cai JP. 8-Oxoguanine modifications in peripheral leukocyte microRNAs promote age-related inflammation by remodeling gene regulatory networks. Free Radic Biol Med 2025; 235:261-279. [PMID: 40288701 DOI: 10.1016/j.freeradbiomed.2025.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
The role of reactive oxygen species (ROS) in cellular senescence and inflammation has been extensively studied; however, the specific molecular mechanisms underlying these effects have yet to be fully elucidated. By applying a nucleotide pool detection system, oxidative microRNA sequencing, and cytokine chip detection techniques, we found that the levels of 8-oxo-guanosine triphosphate (8-oxo-GTP) increased with age and were positively correlated with elevated levels of oxidized microRNA and increased levels of inflammatory factors. During aging, guanine residues in the seed regions of miR-98-5p that regulates ICAM1, and miR-8085 that regulates CXCL16, were oxidized to 8-oxoguanine. This oxidation weakened the binding affinity of microRNAs to their target genes, thereby promoting the expression of ICAM1 and CXCL16. Oxidized microRNAs can regulate new target genes via o8G:A base mismatches. Our findings reveal that ROS-induced oxidative modifications of microRNAs reshape gene regulatory networks, activate inflammatory pathways, providing new insights into the mechanisms of age-related inflammation.
Collapse
Affiliation(s)
- Ming-Ming Han
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Department of Clinical Laboratory, Aerospace Center Hospital, Beijing 100049, China
| | - Lan Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Jin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Qing-Yu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ying-Min Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Jia-Qi Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lv-Tao Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Ruo-Mei Qi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [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: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
4
|
Ma L, Meng Y, An Y, Han P, Zhang C, Yue Y, Wen C, Shi X, Jin J, Yang G, Li X. Single-cell RNA-seq reveals novel interaction between muscle satellite cells and fibro-adipogenic progenitors mediated with FGF7 signalling. J Cachexia Sarcopenia Muscle 2024; 15:1388-1403. [PMID: 38751367 PMCID: PMC11294021 DOI: 10.1002/jcsm.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Muscle satellite cells (MuSCs) exert essential roles in skeletal muscle adaptation to growth, injury and ageing, and their functions are extensively modulated by microenvironmental factors. However, the current knowledge about the interaction of MuSCs with niche cells is quite limited. METHODS A 10× single-cell RNA sequencing (scRNA-seq) was performed on porcine longissimus dorsi and soleus (SOL) muscles to generate a single-cell transcriptomic dataset of myogenic cells and other cell types. Sophisticated bioinformatic analyses, including unsupervised clustering analysis, marker gene, gene set variation analysis (GSVA), AUCell, pseudotime analysis and RNA velocity analysis, were performed to explore the heterogeneity of myogenic cells. CellChat analysis was used to demonstrate cell-cell communications across myogenic cell subpopulations and niche cells, especially fibro-adipogenic progenitors (FAPs). Integrated analysis with human and mice datasets was performed to verify the expression of FGF7 across diverse species. The role of FGF7 on MuSC proliferation was evaluated through administering recombinant FGF7 to porcine MuSCs, C2C12, cardiotoxin (CTX)-injured muscle and d-galactose (d-gal)-induced ageing model. RESULTS ScRNA-seq totally figured out five cell types including myo-lineage cells and FAPs, and myo-lineage cells were further classified into six subpopulations, termed as RCN3+, S100A4+, ID3+, cycling (MKI67+), MYF6+ and MYMK+ satellite cells, respectively. There was a higher proportion of cycling and MYF6+ cells in the SOL population. CellChat analysis uncovered a particular impact of FAPs on myogenic cells mediated by FGF7, which was relatively highly expressed in SOL samples. Administration of FGF7 (10 ng/mL) significantly increased the proportion of EdU+ porcine MuSCs and C2C12 by 4.03 ± 0.81% (P < 0.01) and 6.87 ± 2.17% (P < 0.05), respectively, and knockdown of FGFR2 dramatically abolished the pro-proliferating effects (P < 0.05). In CTX-injured muscle, FGF7 significantly increased the ratio of EdU+/Pax7+ cells by 15.68 ± 5.45% (P < 0.05) and elevated the number of eMyHC+ regenerating myofibres by 19.7 ± 4.25% (P < 0.01). Under d-gal stimuli, FGF7 significantly reduced γH2AX+ cells by 17.19 ± 3.05% (P < 0.01) in porcine MuSCs, induced EdU+ cells by 4.34 ± 1.54% (P < 0.05) in C2C12, and restored myofibre size loss and running exhaustion in vivo (all P < 0.05). CONCLUSIONS Our scRNA-seq reveals a novel interaction between muscle FAPs and satellite cells mediated by FGF7-FGFR2. Exogenous FGF7 augments the proliferation of satellite cells and thus benefits muscle regeneration and counteracts age-related myopathy.
Collapse
Affiliation(s)
- Lu Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Yingying Meng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Yalong An
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Peiyuan Han
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Chen Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Yongqi Yue
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Chenglong Wen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Xin'e Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Jianjun Jin
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| | - Xiao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityXianyangChina
| |
Collapse
|
5
|
Liu F, Cao Y, Wang X, Zhang K, Li N, Su Y, Zhang Y, Meng Q. Islr regulates satellite cells asymmetric division through the SPARC/p-ERK1/2 signaling pathway. FASEB J 2024; 38:e23534. [PMID: 38597911 DOI: 10.1096/fj.202302614r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 04/11/2024]
Abstract
Satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute and chronic muscle injuries. The balance between stem cell self-renewal and differentiation determines the kinetics and efficiency of skeletal muscle regeneration. This study assessed the function of Islr in SC asymmetric division. The deletion of Islr reduced muscle regeneration in adult mice by decreasing the SC pool. Islr is pivotal for SC proliferation, and its deletion promoted the asymmetric division of SCs. A mechanistic search revealed that Islr bound to and degraded secreted protein acidic and rich in cysteine (SPARC), which activated p-ERK1/2 signaling required for asymmetric division. These findings demonstrate that Islr is a key regulator of SC division through the SPARC/p-ERK1/2 signaling pathway. These data provide a basis for treating myopathy.
Collapse
Affiliation(s)
- Fan Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yuxin Cao
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Xiong Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kuo Zhang
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Na Li
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yang Su
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qingyong Meng
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| |
Collapse
|
6
|
Kim SJ, Kwon S, Chung S, Lee EJ, Park SE, Choi SJ, Oh SY, Ryu GH, Jeon HB, Chang JW. Nervonic Acid Inhibits Replicative Senescence of Human Wharton's Jelly-Derived Mesenchymal Stem Cells. Int J Stem Cells 2024; 17:80-90. [PMID: 37822280 PMCID: PMC10899888 DOI: 10.15283/ijsc23101] [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: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023] Open
Abstract
Cellular senescence causes cell cycle arrest and promotes permanent cessation of proliferation. Since the senescence of mesenchymal stem cells (MSCs) reduces proliferation and multipotency and increases immunogenicity, aged MSCs are not suitable for cell therapy. Therefore, it is important to inhibit cellular senescence in MSCs. It has recently been reported that metabolites can control aging diseases. Therefore, we aimed to identify novel metabolites that regulate the replicative senescence in MSCs. Using a fecal metabolites library, we identified nervonic acid (NA) as a candidate metabolite for replicative senescence regulation. In replicative senescent MSCs, NA reduced senescence-associated β-galactosidase positive cells, the expression of senescence-related genes, as well as increased stemness and adipogenesis. Moreover, in non-senescent MSCs, NA treatment delayed senescence caused by sequential subculture and promoted proliferation. We confirmed, for the first time, that NA delayed and inhibited cellular senescence. Considering optimal concentration, duration, and timing of drug treatment, NA is a novel potential metabolite that can be used in the development of technologies that regulate cellular senescence.
Collapse
Affiliation(s)
- Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Soojin Kwon
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Soobeen Chung
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Eun Joo Lee
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Korea
| | - Gyu Ha Ryu
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea
- The Office of R&D Strategy & Planning, Samsung Medical Center, Seoul, Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
7
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement. ACS Chem Neurosci 2024; 15:1-30. [PMID: 38095562 PMCID: PMC10767750 DOI: 10.1021/acschemneuro.3c00531] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024] Open
Abstract
Aging is a dynamic, time-dependent process that is characterized by a gradual accumulation of cell damage. Continual functional decline in the intrinsic ability of living organisms to accurately regulate homeostasis leads to increased susceptibility and vulnerability to diseases. Many efforts have been put forth to understand and prevent the effects of aging. Thus, the major cellular and molecular hallmarks of aging have been identified, and their relationships to age-related diseases and malfunctions have been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent aging-related research. We review the advances in knowledge and delineate trends in research advancements on aging factors and attributes across time and geography. We also review the current concepts related to the major aging hallmarks on the molecular, cellular, and organismic level, age-associated diseases, with attention to brain aging and brain health, as well as the major biochemical processes associated with aging. Major age-related diseases have been outlined, and their correlations with the major aging features and attributes are explored. We hope this review will be helpful for apprehending the current knowledge in the field of aging mechanisms and progression, in an effort to further solve the remaining challenges and fulfill its potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
8
|
Liu W, Wang T, Wang W, Lin X, Xie K. Tanshinone IIA promotes the proliferation and differentiation ability of primary muscle stem cells via MAPK and Akt signaling. Biochem Biophys Res Commun 2023; 689:149235. [PMID: 37976834 DOI: 10.1016/j.bbrc.2023.149235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Salvia miltiorrhiza Bunge is a widely-used traditional Chinese medicine to treat a variety of diseases including muscle disorders. The underlying pharmacological mechanisms of which active component and how it functions are still unknown. Tanshinone IIA (Tan IIA) is the main active lipophilic compound in Salvia miltiorrhiza Bunge. Muscle stem cells (MuSCs) play a crucial role in maintaining healthy physiological function of skeletal muscle. For the purpose of this study, we investigated the effects of Tan IIA on primary MuSCs as well as mechanism. The EdU staining, cell counts assay and RT-qPCR results of proliferative genes revealed increased proliferation ability of MuSCs after Tan IIA treatment. Immunofluorescent staining of MyHC and RT-qPCR results of myogenic genes found Tan IIA contributed to promoting differentiation of MuSCs. In addition, enrichment analysis of RNA-seq data and Western blot assay results demonstrated activated MAPK and Akt signaling after treatment of Tan IIA during proliferation and differentiation. The above proliferative and differentiative phonotypes could be suppressed by the combination of MAPK inhibitor U0126 and Akt inhibitor Akti 1/2, respectively. Furthermore, HE staining found significantly improved myofiber regeneration of injured muscle after Tan IIA treatment, which also contributed to muscle force and running performance recovery. Thus, Tan IIA could promote proliferation and differentiation ability of MuSCs through activating MAPK and Akt signaling, respectively. These beneficial effects also significantly contributed to muscle regeneration and muscle function recovery after muscle injury.
Collapse
Affiliation(s)
- Wenbin Liu
- Department of Orthopedic Surgery, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, No.57 Canghou Street, Wenzhou, Zhejiang, PR China
| | - Tihui Wang
- Department of Orthopedic Surgery, Mindong Hospital Affiliated to Fujian Medical University, No.89 Heshan Road, Fuan, Fujian, PR China
| | - Wei Wang
- Department of Orthopedic Surgery, HuBei Provincial Hospital of TCM, No.4 Hua Yuan Shan, Wuchang District, Wuhan, Hubei, PR China
| | - Xingzuan Lin
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, PR China.
| | - Kailuo Xie
- Department of Orthopedic Surgery, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, No.57 Canghou Street, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
9
|
Lee J, Park J, Choe H, Shim K. Insect peptide CopA3 promotes proliferation and PAX7 and MYOD expression in porcine muscle satellite cells. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1132-1143. [PMID: 36812017 PMCID: PMC9890342 DOI: 10.5187/jast.2022.e81] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Insects are a valuable natural source that can produce a variety of bioactive compounds due to their increasing species diversity. CopA3 is an antimicrobial peptide derived from Copris tripartitus (i.e., the dung beetle). It is known to increase the proliferation of colonic epithelial and neuronal stem cells by regulating cell cycle. This research hypothesized that CopA3 can promote the proliferation of porcine muscle satellite cells (MSCs). The effects of CopA3 on porcine MSCs, which are important for muscle growth and regeneration, remain unclear. Here, we investigated the effects of CopA3 on porcine MSCs. According to viability results, we designed four groups: control (without CopA3) and three treatment groups (treated with 5,10, and 25 μg/mL of CopA3). At a CopA3 concentration of 5 μg/mL and 10 μg/mL, the proliferation of MSCs increased more than that observed in the control group. Furthermore, compared to that in the control, CopA3 treatment increased the S phase but decreased the G0/G1 phase ratio. Additionally, early and late apoptotic cells were found to be decreased in the 5 μg/mL group. The expressions of the myogenesis-related transcription factor PAX7 and MYOD proteins were significantly upregulated in the 5 μg/mL and 10 μg/mL groups, whereas the MYOG protein remained undetected in all group. This study suggested that CopA3 promotes muscle cell proliferation by regulating the cell cycle of MSCs and can regulate the activity of MSCs by increasing the expressions of PAX7 and MYOD.
Collapse
Affiliation(s)
- Jeongeun Lee
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea
| | - Jinryoung Park
- Department of Stem Cell and Regenerative
Biotechnology, Konkuk University, Seoul 06591, Korea,3D Tissue Culture Research Center, Konkuk
University, Seoul 06591, Korea
| | - Hosung Choe
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| | - Kwanseob Shim
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea,Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea,Corresponding author: Kwanseob Shim,
Department of Agricultural Convergence Technology, Jeonbuk National University,
Jeonju 54896, Korea. Tel: +82-63-270-2609, E-mail:
| |
Collapse
|
10
|
From cyclins to CDKIs: Cell cycle regulation of skeletal muscle stem cell quiescence and activation. Exp Cell Res 2022; 420:113275. [PMID: 35931143 DOI: 10.1016/j.yexcr.2022.113275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/12/2022] [Accepted: 07/03/2022] [Indexed: 11/22/2022]
Abstract
After extensive proliferation during development, the adult skeletal muscle cells remain outside the cell cycle, either as post-mitotic myofibers or as quiescent muscle stem cells (MuSCs). Despite its terminally differentiated state, adult skeletal muscle has a remarkable regeneration potential, driven by MuSCs. Upon injury, MuSC quiescence is reversed to support tissue growth and repair and it is re-established after the completion of muscle regeneration. The distinct cell cycle states and transitions observed in the different myogenic populations are orchestrated by elements of the cell cycle machinery. This consists of i) complexes of cyclins and Cyclin-Dependent Kinases (CDKs) that ensure cell cycle progression and ii) their negative regulators, the Cyclin-Dependent Kinase Inhibitors (CDKIs). In this review we discuss the roles of these factors in developmental and adult myogenesis, with a focus on CDKIs that have emerging roles in stem cell functions.
Collapse
|
11
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
12
|
Wang J, Broer T, Chavez T, Zhou CJ, Tran S, Xiang Y, Khodabukus A, Diao Y, Bursac N. Myoblast deactivation within engineered human skeletal muscle creates a transcriptionally heterogeneous population of quiescent satellite-like cells. Biomaterials 2022; 284:121508. [PMID: 35421801 PMCID: PMC9289780 DOI: 10.1016/j.biomaterials.2022.121508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Satellite cells (SCs), the adult Pax7-expressing stem cells of skeletal muscle, are essential for muscle repair. However, in vitro investigations of SC function are challenging due to isolation-induced SC activation, loss of native quiescent state, and differentiation to myoblasts. In the present study, we optimized methods to deactivate in vitro expanded human myoblasts within a 3D culture environment of engineered human skeletal muscle tissues ("myobundles"). Immunostaining and gene expression analyses revealed that a fraction of myoblasts within myobundles adopted a quiescent phenotype (3D-SCs) characterized by increased Pax7 expression, cell cycle exit, and activation of Notch signaling. Similar to native SCs, 3D-SC quiescence is regulated by Notch and Wnt signaling while loss of quiescence and reactivation of 3D-SCs can be induced by growth factors including bFGF. Myobundle injury with a bee toxin, melittin, induces robust myofiber fragmentation, functional decline, and 3D-SC proliferation. By applying single cell RNA-sequencing (scRNA-seq), we discover the existence of two 3D-SC subpopulations (quiescent and activated), identify deactivation-associated gene signature using trajectory inference between 2D myoblasts and 3D-SCs, and characterize the transcriptomic changes within reactivated 3D-SCs in response to melittin-induced injury. These results demonstrate the ability of an in vitro engineered 3D human skeletal muscle environment to support the formation of a quiescent and heterogeneous SC population recapitulating several aspects of the native SC phenotype, and provide a platform for future studies of human muscle regeneration and disease-associated SC dysfunction.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
14
|
Zhou Q, Wang W, Wu J, Qiu S, Yuan S, Fu PL, Qian QR, Xu YZ. Ubiquitin-specific protease 3 attenuates interleukin-1β-mediated chondrocyte senescence by deacetylating forkhead box O-3 via sirtuin-3. Bioengineered 2021; 13:2017-2027. [PMID: 34847835 PMCID: PMC8974216 DOI: 10.1080/21655979.2021.2012552] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Osteoarthritis (OA) affects approximately 12% of the aging Western population. The sirtuin/forkhead box O (SIRT/FOXO) signaling pathway plays essential roles in various biological processes. Despite it has been demonstrated that ubiquitin-specific protease 3 (USP3) inhibits chondrocyte apoptosis induced by interleukin (IL)-1β, the role of USP3/SIRT3/FOXO3 in the senescence of chondrocytes in OA is unclear. This study initially isolated articular chondrocytes and investigated the role of USP3 in IL-1β-induced senescence of chondrocytes. After USP3 was overexpressed or silenced by lentivirus, expressions of genes and proteins were detected using quantitative polymerase chain reaction and immunoblotting, respectively. Cell cycle analysis was performed using flow cytometry. Reactive oxygen species (ROS) levels and senescence were analyzed. Then, SIRT3 was inhibited or overexpressed to explore the underlying mechanism. We found that overexpression of USP3 hindered IL-1β-mediated cell cycle arrest, ROS generation, and chondrocyte senescence. The inhibition of SIRT3 blocked the protective effect of USP3 on cell senescence, whereas the overexpression of SIRT3 abolished USP3-silencing-induced cell senescence. Furthermore, SIRT3 attenuated cell senescence, probably by deacetylating FOXO3. USP3 upregulated SIRT3 to deacetylate FOXO3 and attenuated IL-1β-induced chondrocyte senescence. This study demonstrated that USP3 probably attenuated IL-1β-mediated chondrocyte senescence by deacetylating FOXO3 via SIRT3.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China.,Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Wei Wang
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Jun Wu
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Shang Qiu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, PR China
| | - Shuai Yuan
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Pei-Liang Fu
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Qi-Rong Qian
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Yao-Zeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China
| |
Collapse
|
15
|
Kim JW, Kim R, Choi H, Lee SJ, Bae GU. Understanding of sarcopenia: from definition to therapeutic strategies. Arch Pharm Res 2021; 44:876-889. [PMID: 34537916 DOI: 10.1007/s12272-021-01349-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Sarcopenia refers to the gradual loss of skeletal muscle mass and function along with aging and is a social burden due to growing healthcare cost associated with a super-aging society. Therefore, researchers have established guidelines and tests to diagnose sarcopenia. Several studies have been conducted actively to reveal the cause of sarcopenia and find an economic therapy to improve the quality of life in elderly individuals. Sarcopenia is caused by multiple factors such as reduced regenerative capacity, imbalance in protein turnover, alteration of fat and fibrotic composition in muscle, increased reactive oxygen species, dysfunction of mitochondria and increased inflammation. Based on these mechanisms, nonpharmacological and pharmacological strategies have been developed to prevent and treat sarcopenia. Although several studies are currently in progress, no treatment is available yet. This review presents the definition of sarcopenia and summarizes recent understanding on the detailed mechanisms, diagnostic criteria, and strategies for prevention and treatment.
Collapse
Affiliation(s)
- Jee Won Kim
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ryuni Kim
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Hyerim Choi
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sang-Jin Lee
- Research Institute of Aging-Related Disease, AniMusCure Inc., Suwon, 16419, Republic of Korea.
| | - Gyu-Un Bae
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
16
|
FGF-2-dependent signaling activated in aged human skeletal muscle promotes intramuscular adipogenesis. Proc Natl Acad Sci U S A 2021; 118:2021013118. [PMID: 34493647 PMCID: PMC8449320 DOI: 10.1073/pnas.2021013118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/23/2021] [Indexed: 01/07/2023] Open
Abstract
Aged skeletal muscle is markedly affected by fatty muscle infiltration, and strategies to reduce the occurrence of intramuscular adipocytes are urgently needed. Here, we show that fibroblast growth factor-2 (FGF-2) not only stimulates muscle growth but also promotes intramuscular adipogenesis. Using multiple screening assays upstream and downstream of microRNA (miR)-29a signaling, we located the secreted protein and adipogenic inhibitor SPARC to an FGF-2 signaling pathway that is conserved between skeletal muscle cells from mice and humans and that is activated in skeletal muscle of aged mice and humans. FGF-2 induces the miR-29a/SPARC axis through transcriptional activation of FRA-1, which binds and activates an evolutionary conserved AP-1 site element proximal in the miR-29a promoter. Genetic deletions in muscle cells and adeno-associated virus-mediated overexpression of FGF-2 or SPARC in mouse skeletal muscle revealed that this axis regulates differentiation of fibro/adipogenic progenitors in vitro and intramuscular adipose tissue (IMAT) formation in vivo. Skeletal muscle from human donors aged >75 y versus <55 y showed activation of FGF-2-dependent signaling and increased IMAT. Thus, our data highlights a disparate role of FGF-2 in adult skeletal muscle and reveals a pathway to combat fat accumulation in aged human skeletal muscle.
Collapse
|
17
|
Pi C, Ma C, Wang H, Sun H, Yu X, Gao X, Yang Y, Sun Y, Zhang H, Shi Y, Li Y, Li Y, He X. MiR-34a suppression targets Nampt to ameliorate bone marrow mesenchymal stem cell senescence by regulating NAD +-Sirt1 pathway. Stem Cell Res Ther 2021; 12:271. [PMID: 33957971 PMCID: PMC8101138 DOI: 10.1186/s13287-021-02339-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Expansion-mediated replicative senescence and age-related natural senescence have adverse effects on mesenchymal stem cell (MSC) regenerative capability and functionality, thus severely impairing the extensive applications of MSC-based therapies. Emerging evidences suggest that microRNA-34a (miR-34a) has been implicated in the process of MSC senescence; however, the molecular mechanisms with regard to how miR-34a influencing MSC senescence remain largely undetermined. METHODS MiR-34a expression in MSCs was evaluated utilizing RT-qPCR. The functional effects of miR-34a exerting on MSC senescence were investigated via gene manipulation. Relevant gene and protein expression levels were analyzed by RT-qPCR and western blot. Luciferase reporter assays were applied to confirm that Nampt is a direct target of miR-34a. The underlying regulatory mechanism of miR-34a targeting Nampt in MSC senescence was further explored by measuring intracellular NAD+ content, NAD+/NADH ratio and Sirt1 activity. RESULTS In contrast to Nampt expression, miR-34a expression incremented in senescent MSCs. MiR-34a overexpression in young MSCs resulted in senescence-associated characteristics as displayed by senescence-like morphology, prolonged cell proliferation, declined osteogenic differentiation potency, heightened senescence-associated-β-galactosidase activity, and upregulated expression levels of the senescence-associated factors. Conversely, miR-34a suppression in replicative senescent and natural senescent MSCs contributed to diminished senescence-related phenotypic features. We identified Nampt as a direct target gene of miR-34a. In addition, miR-34a repletion resulted in prominent reductions in Nampt expression levels, NAD+ content, NAD+/NADH ratio, and Sirt1 activity, whereas anti-miR-34a treatment exerted the opposite effects. Furthermore, miR-34a-mediated MSC senescence was evidently rescued following the co-treatment with Nampt overexpression. CONCLUSION This study identifies a significant role of miR-34a playing in MSC replicative senescence and natural senescence via targeting Nampt and further mediating by NAD+-Sirt1 pathway, carrying great implications for optimal strategies for MSC therapeutic applications.
Collapse
Affiliation(s)
- Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China.,The First Hospital, and Institute of Immunology, Jilin University, Changchun, 130021, China
| | - Cao Ma
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Huan Wang
- Department of Pathology, The First Affiliated Hospital, Henan University of Chinese Medicine, Henan, 450000, China
| | - Hui Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xiao Yu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xingyu Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yue Yang
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yanan Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Haiying Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China.
| |
Collapse
|
18
|
Gudagudi KB, Myburgh KH. Methods to Mimic In Vivo Muscle Cell Biology in Primary Human Myoblasts Using Quiescence as a Guidepost in Regenerative Medicine Research. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:176-189. [PMID: 33635139 DOI: 10.1089/omi.2020.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Regenerative medicine research and testing of new therapeutics for muscle-related human diseases call for a deeper understanding of how human myoblasts gain and maintain quiescence in vitro versus in vivo. The more closely we can experimentally simulate the in vivo environment, the more relevance in vitro research on myoblasts will have. In this context, isolation of satellite cells from muscle tissue causes activation while myoblasts remain activated in culture, thus not simulating quiescence as in their in vivo niche. Cells synchronized for cell cycle present a good starting point for experimental intervention. In the past, myoblast quiescence has been induced using suspension culture (SuCu) and, recently, by knockout serum replacement (KOSR)-supplemented culture media. We assessed the proportion of cells in G0 and molecular regulators after combining the two quiescence-inducing approaches. Quiescence was induced in primary human myoblasts (PHMs) in vitro using KOSR-treatment for 10 days or suspension in viscous media for 2 days (SuCu), or suspension combined with KOSR-treatment for 2 days (blended method, SuCu-KOSR). Quiescence and synchronization were achieved with all three protocols (G0/G1 cell cycle arrest >90% cells). Fold-change of cell cycle controller p21 mRNA for KOSR and SuCu was 3.23 ± 0.30 and 2.86 ± 0.15, respectively. Since this was already a significant change (p < 0.05), no further change was gained with the blended method. But SuCu-KOSR significantly decreased Ki67 (p = 0.0019). Myogenic regulatory factors, Myf5 and MyoD gene expression in PHMs were much more suppressed (p = 0.0004 and p = 0.0034, respectively) in SuCu-KOSR, compared to SuCu alone. In conclusion, a homogenous pool of quiescent primary myoblasts synchronized in the G0 cell cycle phase was achieved with cells from three different donors regardless of the experimental protocol. Myogenic dedifferentiation at the level of Myogenic Regulatory Factors was greater when exposed to the blend of suspension and serum-free culture. We suggest that this blended new protocol can be considered in future biomedical research if differentiation is detected too early during myoblast expansion. This shall also inform new ways to bridge the in vitro and in vivo divides in regenerative medicine research.
Collapse
Affiliation(s)
- Kirankumar B Gudagudi
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Kathryn H Myburgh
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
19
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 473] [Impact Index Per Article: 94.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
20
|
LRTM1 promotes the differentiation of myoblast cells by negatively regulating the FGFR1 signaling pathway. Exp Cell Res 2020; 396:112237. [PMID: 32841643 DOI: 10.1016/j.yexcr.2020.112237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 11/23/2022]
Abstract
The proliferation and differentiation of myoblast cells are regulated by the fibroblast growth factor receptor (FGFR) signaling pathway. Although the regulation of FGFR signaling cascades has been widely investigated, the inhibitory mechanism that particularly function in skeletal muscle myogenesis remains obscure. In this study, we determined that LRTM1, an inhibitory regulator of the FGFR signaling pathway, negatively modulates the activation of ERK and promotes the differentiation of myoblast cells. LRTM1 is dynamically expressed during myoblast differentiation and skeletal muscle regeneration after injury. In mouse myoblast C2C12 cells, knockout (KO) of Lrtm1 significantly prevents the differentiation of myoblast cells; this effect is associated with the reduction of MyoD transcriptional activity and the overactivation of ERK kinase. Notably, further studies demonstrated that LRTM1 associates with p52Shc and inhibits the recruitment of p52Shc to FGFR1. Taken together, our findings identify a novel negative regulator of FGFR1, which plays an important role in regulating the differentiation of myoblast cells.
Collapse
|
21
|
Abstract
Adult stem cells undergo both replicative and chronological aging in their niches, with catastrophic declines in regenerative potential with age. Due to repeated environmental insults during aging, the chromatin landscape of stem cells erodes, with changes in both DNA and histone modifications, accumulation of damage, and altered transcriptional response. A body of work has shown that altered chromatin is a driver of cell fate changes and a regulator of self-renewal in stem cells and therefore a prime target for juvenescence therapeutics. This review focuses on chromatin changes in stem cell aging and provides a composite view of both common and unique epigenetic themes apparent from the studies of multiple stem cell types.
Collapse
Affiliation(s)
- Changyou Shi
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Lin Wang
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
22
|
Pi C, Yang Y, Sun Y, Wang H, Sun H, Ma M, Lin L, Shi Y, Li Y, Li Y, He X. Nicotinamide phosphoribosyltransferase postpones rat bone marrow mesenchymal stem cell senescence by mediating NAD +-Sirt1 signaling. Aging (Albany NY) 2020; 11:3505-3522. [PMID: 31175267 PMCID: PMC6594813 DOI: 10.18632/aging.101993] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/22/2019] [Indexed: 12/25/2022]
Abstract
In vitro replicative senescence affects MSC characteristics and functionality, thus severely restricting their application in regenerative medicine and MSC-based therapies. Previously, we found that MSC natural senescence is accompanied by altered intracellular nicotinamide adenine dinucleotide (NAD+) metabolism, in which Nampt plays a key role. However, whether Nampt influences MSC replicative senescence is still unclear. Our study showed that Nampt expression is down-regulated during MSC replicative senescence. Nampt depletion via a specific Nampt inhibitor FK866 or Nampt knockdown in early passage MSCs led to enhanced senescence as indicated by senescence-like morphology, reduced proliferation, and adipogenic and osteogenic differentiation, and increased senescence-associated-β-galactosidase activity and the expression of the senescence-associated factor p16INK4a. Conversely, Nampt overexpression ameliorated senescence-associated phenotypic features in late passage MSCs. Further, Nampt inhibition resulted in reduced intracellular NAD+ content, NAD+/NADH ratio, and Sirt1 activity, whereas overexpression had the opposite effects. Exogenous intermediates involved in NAD+ biosynthesis not only rescued replicative senescent MSCs but also alleviated FK866-induced MSC senescence. Thus, Nampt suppresses MSC senescence via mediating NAD+-Sirt1 signaling. This study provides novel mechanistic insights into MSC replicative senescence and a promising strategy for the severe shortage of cells for MSC-based therapies.
Collapse
Affiliation(s)
- Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,The First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yue Yang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Yanan Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Huan Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Hui Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Mao Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lin Lin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
23
|
Gudagudi KB, d’Entrèves NP, Woudberg NJ, Steyn PJ, Myburgh KH. In vitro induction of quiescence in isolated primary human myoblasts. Cytotechnology 2020; 72:189-202. [PMID: 31993891 PMCID: PMC7192999 DOI: 10.1007/s10616-019-00365-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Adult skeletal muscle stem cells, satellite cells, remain in an inactive or quiescent state in vivo under physiological conditions. Progression through the cell cycle, including activation of quiescent cells, is a tightly regulated process. Studies employing in vitro culture of satellite cells, primary human myoblasts (PHMs), necessitate isolation myoblasts from muscle biopsies. Further studies utilizing these cells should endeavour to represent their native in vivo characteristics as closely as possible, also considering variability between individual donors. This study demonstrates the approach of utilizing KnockOut™ Serum Replacement (KOSR)-supplemented culture media as a quiescence-induction media for 10 days in PHMs isolated and expanded from three different donors. Cell cycle analysis demonstrated that treatment resulted in an increase in G1 phase and decreased S phase proportions in all donors (p < 0.005). The proportions of cells in G1 and G2 phases differed in proliferating myoblasts when comparing donors (p < 0.05 to p < 0.005), but following KOSR treatment, the proportion of cells in G1 (p = 0.558), S (p = 0.606) and G2 phases (p = 0.884) were equivalent between donors. When cultured in the quiescence-induction media, expression of CD34 and Myf5 remained constant above > 98% over time from day 0 to day 10. In contrast activation (CD56), proliferation (Ki67) and myogenic marker MyoD decreased, indicated de-differentiation. Induction of quiescence was accompanied in all three clones by fold change in p21 mRNA greater than 3.5 and up to tenfold. After induction of quiescence, differentiation into myotubes was not affected. In conclusion, we describe a method to induce quiescence in PHMs from different donors.
Collapse
Affiliation(s)
- Kirankumar B. Gudagudi
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Niccolò Passerin d’Entrèves
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Nicholas J. Woudberg
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Paul J. Steyn
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
- Department of Human Biology, University of Cape Town, Anzio Road, Observatory, South Africa
| | - Kathryn H. Myburgh
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| |
Collapse
|
24
|
Falah G, Giller A, Gutman D, Atzmon G. Breaking the Glass Ceiling. Gerontology 2020; 66:309-314. [PMID: 32101855 DOI: 10.1159/000505995] [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: 07/04/2019] [Accepted: 01/17/2020] [Indexed: 11/19/2022] Open
Abstract
Over the past century, the life expectancy in industrialized countries has rapidly risen by over 30 years due to improvements in standards of medical care, sanitation, and lifestyle. Estimation of life expectancy has traditionally been viewed through a lens of epidemiology and public health. However, this data, while considered the "gold standard" of measuring healthy life expectancy, may soon find itself redundant in the face of advancing medical technology. Even as average life expectancy has increased, there has not been an equivalent increase in healthy life expectancy, or "healthspan"; furthermore, there is a current trend of stagnation in life expectancy, as the supposed increases are estimated to be drastically slowing down, in part due to exhaustion of our current ability to extend the human lifespan. In this viewpoint, we will examine the developing fields of medicine and life sciences which will reshape our current approach to life expectancy prediction.
Collapse
Affiliation(s)
- Ghadeer Falah
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Abram Giller
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Danielle Gutman
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Gil Atzmon
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel,
| |
Collapse
|
25
|
Joanisse S, Lim C, McKendry J, Mcleod JC, Stokes T, Phillips SM. Recent advances in understanding resistance exercise training-induced skeletal muscle hypertrophy in humans. F1000Res 2020; 9. [PMID: 32148775 PMCID: PMC7043134 DOI: 10.12688/f1000research.21588.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/18/2020] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle plays a pivotal role in the maintenance of physical and metabolic health and, critically, mobility. Accordingly, strategies focused on increasing the quality and quantity of skeletal muscle are relevant, and resistance exercise is foundational to the process of functional hypertrophy. Much of our current understanding of skeletal muscle hypertrophy can be attributed to the development and utilization of stable isotopically labeled tracers. We know that resistance exercise and sufficient protein intake act synergistically and provide the most effective stimuli to enhance skeletal muscle mass; however, the molecular intricacies that underpin the tremendous response variability to resistance exercise-induced hypertrophy are complex. The purpose of this review is to discuss recent studies with the aim of shedding light on key regulatory mechanisms that dictate hypertrophic gains in skeletal muscle mass. We also aim to provide a brief up-to-date summary of the recent advances in our understanding of skeletal muscle hypertrophy in response to resistance training in humans.
Collapse
Affiliation(s)
- Sophie Joanisse
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Changhyun Lim
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - James McKendry
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Jonathan C Mcleod
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Tanner Stokes
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
26
|
Etienne J, Liu C, Skinner CM, Conboy MJ, Conboy IM. Skeletal muscle as an experimental model of choice to study tissue aging and rejuvenation. Skelet Muscle 2020; 10:4. [PMID: 32033591 PMCID: PMC7007696 DOI: 10.1186/s13395-020-0222-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle is among the most age-sensitive tissues in mammal organisms. Significant changes in its resident stem cells (i.e., satellite cells, SCs), differentiated cells (i.e., myofibers), and extracellular matrix cause a decline in tissue homeostasis, function, and regenerative capacity. Based on the conservation of aging across tissues and taking advantage of the relatively well-characterization of the myofibers and associated SCs, skeletal muscle emerged as an experimental system to study the decline in function and maintenance of old tissues and to explore rejuvenation strategies. In this review, we summarize the approaches for understanding the aging process and for assaying the success of rejuvenation that use skeletal muscle as the experimental system of choice. We further discuss (and exemplify with studies of skeletal muscle) how conflicting results might be due to variations in the techniques of stem cell isolation, differences in the assays of functional rejuvenation, or deciding on the numbers of replicates and experimental cohorts.
Collapse
Affiliation(s)
- Jessy Etienne
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Colin M Skinner
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA.
| |
Collapse
|
27
|
Mehdipour M, Etienne J, Chen CC, Gathwala R, Rehman M, Kato C, Liu C, Liu Y, Zuo Y, Conboy MJ, Conboy IM. Rejuvenation of brain, liver and muscle by simultaneous pharmacological modulation of two signaling determinants, that change in opposite directions with age. Aging (Albany NY) 2019; 11:5628-5645. [PMID: 31422380 PMCID: PMC6710051 DOI: 10.18632/aging.102148] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
We hypothesize that altered intensities of a few morphogenic pathways account for most/all the phenotypes of aging. Investigating this has revealed a novel approach to rejuvenate multiple mammalian tissues by defined pharmacology. Specifically, we pursued the simultaneous youthful in vivo calibration of two determinants: TGF-beta which activates ALK5/pSmad 2,3 and goes up with age, and oxytocin (OT) which activates MAPK and diminishes with age. The dose of Alk5 inhibitor (Alk5i) was reduced by 10-fold and the duration of treatment was shortened (to minimize overt skewing of cell-signaling pathways), yet the positive outcomes were broadened, as compared with our previous studies. Alk5i plus OT quickly and robustly enhanced neurogenesis, reduced neuro-inflammation, improved cognitive performance, and rejuvenated livers and muscle in old mice. Interestingly, the combination also diminished the numbers of cells that express the CDK inhibitor and marker of senescence p16 in vivo. Summarily, simultaneously re-normalizing two pathways that change with age in opposite ways (up vs. down) synergistically reverses multiple symptoms of aging.
Collapse
Affiliation(s)
- Melod Mehdipour
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jessy Etienne
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chia-Chien Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ranveer Gathwala
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Maryam Rehman
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cameron Kato
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yutong Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
28
|
Jia Z, Nie Y, Yue F, Kong Y, Gu L, Gavin TP, Liu X, Kuang S. A requirement of Polo-like kinase 1 in murine embryonic myogenesis and adult muscle regeneration. eLife 2019; 8:e47097. [PMID: 31393265 PMCID: PMC6687435 DOI: 10.7554/elife.47097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/17/2019] [Indexed: 01/22/2023] Open
Abstract
Muscle development and regeneration require delicate cell cycle regulation of embryonic myoblasts and adult muscle satellite cells (MuSCs). Through analysis of the Polo-like kinase (Plk) family cell-cycle regulators in mice, we show that Plk1's expression closely mirrors myoblast dynamics during embryonic and postnatal myogenesis. Cell-specific deletion of Plk1 in embryonic myoblasts leads to depletion of myoblasts, developmental failure and prenatal lethality. Postnatal deletion of Plk1 in MuSCs does not perturb their quiescence but depletes activated MuSCs as they enter the cell cycle, leading to regenerative failure. The Plk1-null MuSCs are arrested at the M-phase, accumulate DNA damage, and apoptose. Mechanistically, Plk1 deletion upregulates p53, and inhibition of p53 promotes survival of the Plk1-null myoblasts. Pharmacological inhibition of Plk1 similarly inhibits proliferation but promotes differentiation of myoblasts in vitro, and blocks muscle regeneration in vivo. These results reveal for the first time an indispensable role of Plk1 in developmental and regenerative myogenesis.
Collapse
Affiliation(s)
- Zhihao Jia
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yaohui Nie
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Feng Yue
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yifan Kong
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Lijie Gu
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Timothy P Gavin
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Xiaoqi Liu
- Department of BiochemistryPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| | - Shihuan Kuang
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| |
Collapse
|
29
|
Chen L, He PL, Yang J, Yang YF, Wang K, Amend B, Stenzl A, Zhang YM, Wang ZL, Xing SS, Luo X. NLRP3/IL1β inflammasome associated with the aging bladder triggers bladder dysfunction in female rats. Mol Med Rep 2019; 19:2960-2968. [PMID: 30720125 PMCID: PMC6423574 DOI: 10.3892/mmr.2019.9919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022] Open
Abstract
Bladder dysfunction is associated with fibrosis-mediated aging, but the corresponding mechanism remains to be elucidated. Activation of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is related to chronic diseases associated with aging, including organ fibrosis. The present study aimed to explore the role of NLRP3/interleukin 1β in aging-associated bladder dysfunction. Female Sprague-Dawley rats were divided into the following two groups (n=10 rats/group): 2-month-old group (young group) and 24-month-old group (old group). Urodynamics were performed to assess the bladder function of the rats. The histological alterations were identified using Masson's trichrome staining. The protein expression of the NLRP3 inflammasome and NAD-dependent protein deacetylase sirtuin-3, mitochondrial (SIRT3) were detected by western blot analysis, and immunohistochemistry was used to examine a senescence marker (p21) and the NLRP3 inflammasome in the bladder. The localization of the key molecule Caspase1 was determined using immunofluorescence. The voiding time was longer in the old group compared with the young group. The expression levels of SIRT3 were reduced in the bladders of the old group, while those of the NLRP3 inflammasome and the senescence marker were significantly higher in the bladders of the old group compared with the young group. Increased collagen deposition leads to chronic bladder fibrosis with increased NLRP3. In the histological examination, the bladders of the old group displayed increased collagen deposition, urothelial thinning and detrusor shrinkage compared with the young group. Tissue fibrosis and urothelial alterations are the principal causes of bladder dysfunction during aging. Downregulated SIRT3 and upregulated expression of the NLRP3 inflammasome are involved in the degradation of aging bladders. Inflamm-aging is a novel mechanism underlying bladder dysfunction.
Collapse
Affiliation(s)
- Lin Chen
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Ping-Lin He
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Jin Yang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Ya-Fei Yang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Kai Wang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Bastian Amend
- Department of Urology, University of Tübingen, D-72074 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen, D-72074 Tübingen, Germany
| | - Ya-Mei Zhang
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Zi-Li Wang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Sha-Sha Xing
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xu Luo
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
30
|
Epigenetic Erosion in Adult Stem Cells: Drivers and Passengers of Aging. Cells 2018; 7:cells7120237. [PMID: 30501028 PMCID: PMC6316114 DOI: 10.3390/cells7120237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
In complex organisms, stem cells are key for tissue maintenance and regeneration. Adult stem cells replenish continuously dividing tissues of the epithelial and connective types, whereas in non-growing muscle and nervous tissues, they are mainly activated upon injury or stress. In addition to replacing deteriorated cells, adult stem cells have to prevent their exhaustion by self-renewal. There is mounting evidence that both differentiation and self-renewal are impaired upon aging, leading to tissue degeneration and functional decline. Understanding the molecular pathways that become deregulate in old stem cells is crucial to counteract aging-associated tissue impairment. In this review, we focus on the epigenetic mechanisms governing the transition between quiescent and active states, as well as the decision between self-renewal and differentiation in three different stem cell types, i.e., spermatogonial stem cells, hematopoietic stem cells, and muscle stem cells. We discuss the epigenetic events that channel stem cell fate decisions, how this epigenetic regulation is altered with age, and how this can lead to tissue dysfunction and disease. Finally, we provide short prospects of strategies to preserve stem cell function and thus promote healthy aging.
Collapse
|
31
|
Boers HE, Haroon M, Le Grand F, Bakker AD, Klein‐Nulend J, Jaspers RT. ---Mechanosensitivity of aged muscle stem cells. J Orthop Res 2018; 36:632-641. [PMID: 29094772 PMCID: PMC5888196 DOI: 10.1002/jor.23797] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/13/2017] [Indexed: 02/04/2023]
Abstract
During aging, skeletal muscle tissue progressively declines in mass, strength, and regenerative capacity. Decreased muscle stem cell (MuSC) number and impaired function might underlie the aging-related muscle wasting and impaired regenerative capacity. As yet, the search for factors that regulate MuSC fate and function has revealed several biochemical factors within the MuSC niche that may be responsible for the decline in MuSC regenerative capacity. This decline cannot be explained by environmental factors solely, as the MuSC potential to regenerate muscle tissue is not reversed by changing the biochemical MuSC niche composition. Here we discuss the likeliness that during physical exercise, MuSCs within their niche are subjected to mechanical loads, in particular pressure and shear stress, as well as associated deformations. We postulate that these physical cues are involved in the activation and differentiation of MuSCs as these cells contain several transmembrane sensor proteins that have been shown to be mechanosensitive in other cell types, that is, endothelial cells and osteoprogenitors. We will specifically address age-related changes in mechanosensing in MuSCs and their niche. Insight in the physical cues applied to the MuSCs in vivo, and how these cues affect MuSC fate and function, helps to develop new therapeutic interventions to counterbalance age-related muscle loss. This requires an approach combining two- and three-dimensional live cell imaging of MuSCs within contracting muscle tissue, mathematical finite element modeling, and cell biology. © 2017 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. on behalf of the Orthopaedic Research Society. J Orthop Res 36:632-641, 2018.
Collapse
Affiliation(s)
- Heleen E. Boers
- Laboratory for MyologyFaculty of Behavioural and Movement SciencesVrije Universiteit AmsterdamAmsterdam Movement SciencesDe Boelelaan 11081081 HZ AmsterdamThe Netherlands
| | - Mohammad Haroon
- Laboratory for MyologyFaculty of Behavioural and Movement SciencesVrije Universiteit AmsterdamAmsterdam Movement SciencesDe Boelelaan 11081081 HZ AmsterdamThe Netherlands
| | - Fabien Le Grand
- Sorbonne UniversitésUPMC Univ Paris 06INSERM UMRS974CNRS FRE3617Center for Research in Myology75013 ParisFrance
| | - Astrid D. Bakker
- Department of Oral Cell BiologyAcademic Centre for Dentistry AmsterdamUniversity of Amsterdam and Vrije Universiteit AmsterdamAmsterdam Movement SciencesAmsterdamThe Netherlands
| | - Jenneke Klein‐Nulend
- Department of Oral Cell BiologyAcademic Centre for Dentistry AmsterdamUniversity of Amsterdam and Vrije Universiteit AmsterdamAmsterdam Movement SciencesAmsterdamThe Netherlands
| | - Richard T. Jaspers
- Laboratory for MyologyFaculty of Behavioural and Movement SciencesVrije Universiteit AmsterdamAmsterdam Movement SciencesDe Boelelaan 11081081 HZ AmsterdamThe Netherlands
| |
Collapse
|
32
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
33
|
Hekmatimoghaddam S, Dehghani Firoozabadi A, Zare-Khormizi MR, Pourrajab F. Sirt1 and Parp1 as epigenome safeguards and microRNAs as SASP-associated signals, in cellular senescence and aging. Ageing Res Rev 2017; 40:120-141. [PMID: 28993289 DOI: 10.1016/j.arr.2017.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/25/2017] [Accepted: 10/05/2017] [Indexed: 01/25/2023]
Abstract
Cellular senescence (CS) is underlying mechanism of organism aging and is closely interconnected with age-related diseases (ARDs). Thus, any attempt that influences CS, may be undertaken to reverse or inhibit senescence, whereby could prolong healthy life span. Until now, two main proposes are epigenetic and genetic modifications of cell fate. The first one concerns rejuvenation through effective reprogramming in cells undergoing senescence, or derived from very old or progeroid patients, by which is effective in vitro in induced pluripotent stem cells (iPSCs). The second approach concerns modification of senescence signaling pathways like as IGF-induced agents. However, senescence research has experienced an unprecedented advance over recent years, particularly with the discovery that the rate of senescence is controlled, at least to some extent, by epigenetic pathways and biochemical processes conserved in evolution. In this review we try to concentrate on very specific pathways (DNA damage response, DDR, and epigenetic modifiers) and very specific determinants (senescence-associated secretory phenotype, SASP-miRNAs) of human premature aging. A major challenge is to dissect the interconnectedness between the candidate elements and their relative contributions to aging, with the final goal of identifying new opportunities for design of novel anti-aging treatments or avoidance of age-associated manifestations. While knowing that aging is unavoidable and we cannot expect its elimination, but prolonging healthy life span is a goal worth serious consideration.
Collapse
Affiliation(s)
- Seyedhossein Hekmatimoghaddam
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Laboratory Sciences, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | | | - Fatemeh Pourrajab
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
34
|
Application of bio-orthogonal proteome labeling to cell transplantation and heterochronic parabiosis. Nat Commun 2017; 8:643. [PMID: 28935952 PMCID: PMC5608760 DOI: 10.1038/s41467-017-00698-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
Studies of heterochronic parabiosis demonstrated that with age, the composition of the circulatory milieu changes in ways that broadly inhibit tissue regenerative capacity. In addition, local tissue niches have age-specific influences on their resident stem cells. Here we use bio-orthogonal proteome labeling for detecting in vivo proteins present only in transplanted myoblasts, but not in host tissue, and proteins exclusive to one young mouse and transferred during parabiosis to its old partner. We use a transgenic mouse strain that ubiquitously expresses a modified tRNA methionine synthase, metRS, which preferentially incorporates the methionine surrogate azido-nor-leucine (ANL) into newly generated proteins. Using click chemistry and a modified antibody array to detect ANL-labeled proteins, we identify several ‘young’ systemic factors in old regenerating muscle of the heterochronic parabiotic partners. Our approach enables the selective profiling of mammalian proteomes in mixed biological environments such as cell and tissue transplantation, apheresis or parabiosis. Clarifying the source of proteins in mixed biological environments, such as after transplantation or parabiosis, remains a challenge. Here, the authors address this need with a mouse strain that incorporates a methionine derivate into proteins, allowing for their detection using click chemistry and antibody arrays.
Collapse
|
35
|
Ghadiali RS, Guimond SE, Turnbull JE, Pisconti A. Dynamic changes in heparan sulfate during muscle differentiation and ageing regulate myoblast cell fate and FGF2 signalling. Matrix Biol 2017; 59:54-68. [PMID: 27496348 PMCID: PMC5380652 DOI: 10.1016/j.matbio.2016.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 01/16/2023]
Abstract
Satellite cells (SCs) are skeletal muscle stem cells residing quiescent around healthy muscle fibres. In response to injury or disease SCs activate, proliferate and eventually differentiate and fuse to one another to form new muscle fibres, or to existing damaged fibres to repair them. The sulfated polysaccharide heparan sulfate (HS) is a highly variable biomolecule known to play key roles in the regulation of cell fate decisions, though the changes that muscle HS undergoes during SC differentiation are unknown. Here we show that the sulfation levels of HS increase during SC differentiation; more specifically, we observe an increase in 6-O and 2-O-sulfation in N-acetylated disaccharides. Interestingly, a specific increase in 6-O sulfation is also observed in the heparanome of ageing muscle, which we show leads to promotion of FGF2 signalling and satellite cell proliferation, suggesting a role for the heparanome dynamics in age-associated loss of quiescence. Addition of HS mimetics to differentiating SC cultures results in differential effects: an oversulfated HS mimetic increases differentiation and inhibits FGF2 signalling, a known major promoter of SC proliferation and inhibitor of differentiation. In contrast, FGF2 signalling is promoted by an N-acetylated HS mimetic, which inhibits differentiation and promotes SC expansion. We conclude that the heparanome of SCs is dynamically regulated during muscle differentiation and ageing, and that such changes might account for some of the phenotypes and signalling events that are associated with these processes.
Collapse
Affiliation(s)
- R S Ghadiali
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - S E Guimond
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - J E Turnbull
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - A Pisconti
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom.
| |
Collapse
|
36
|
Pawlikowski B, Vogler TO, Gadek K, Olwin BB. Regulation of skeletal muscle stem cells by fibroblast growth factors. Dev Dyn 2017; 246:359-367. [PMID: 28249356 DOI: 10.1002/dvdy.24495] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 01/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) are essential for self-renewal of skeletal muscle stem cells (satellite cells) and required for maintenance and repair of skeletal muscle. Satellite cells express high levels of FGF receptors 1 and 4, low levels of FGF receptor 3, and little or no detectable FGF receptor 2. Of the multiple FGFs that influence satellite cell function in culture, FGF2 and FGF6 are the only members that regulate satellite cell function in vivo by activating ERK MAPK, p38α/β MAPKs, PI3 kinase, PLCγ and STATs. Regulation of FGF signaling is complex in satellite cells, requiring Syndecan-4, a heparan sulfate proteoglycan, as well as ß1-integrin and fibronectin. During aging, reduced responsiveness to FGF diminishes satellite cell self-renewal, leading to impaired skeletal muscle regeneration and depletion of satellite cells. Mislocalization of ß1-integrin, reductions in fibronectin, and alterations in heparan sulfate content all contribute to reduced FGF responsiveness in satellite cells. How these cell surface proteins regulate satellite cell self-renewal is incompletely understood. Here we summarize the current knowledge, highlighting the role(s) for FGF signaling in skeletal muscle regeneration, satellite cell behavior, and age-induced muscle wasting. Developmental Dynamics 246:359-367, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bradley Pawlikowski
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Thomas Orion Vogler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Katherine Gadek
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Bradley B Olwin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| |
Collapse
|
37
|
Abstract
This review provides balanced analysis of the advances in systemic regulation of young and old tissue stem cells and suggests strategies for accelerating development of therapies to broadly combat age-related tissue degenerative pathologies. Many highlighted recent reports on systemic tissue rejuvenation combine parabiosis with a “silver bullet” putatively responsible for the positive effects. Attempts to unify these papers reflect the excitement about this experimental approach and add value in reproducing previous work. At the same time, defined molecular approaches, which are “beyond parabiosis” for the rejuvenation of multiple old organs represent progress toward attenuating or even reversing human tissue aging.
Collapse
|
38
|
Chen Y, Pan K, Wang P, Cao Z, Wang W, Wang S, Hu N, Xue J, Li H, Jiang W, Li G, Zhang X. HBP1-mediated Regulation of p21 Protein through the Mdm2/p53 and TCF4/EZH2 Pathways and Its Impact on Cell Senescence and Tumorigenesis. J Biol Chem 2016; 291:12688-12705. [PMID: 27129219 PMCID: PMC4933444 DOI: 10.1074/jbc.m116.714147] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Indexed: 01/09/2023] Open
Abstract
The activity of the CDK inhibitor p21 is associated with diverse biological activities, including cell proliferation, senescence, and tumorigenesis. However, the mechanisms governing transcription of p21 need to be extensively studied. In this study, we demonstrate that the high-mobility group box-containing protein 1 (HBP1) transcription factor is a novel activator of p21 that works as part of a complex mechanism during senescence and tumorigenesis. We found that HBP1 activates the p21 gene through enhancing p53 stability by inhibiting Mdm2-mediated ubiquitination of p53, a well known positive regulator of p21. HBP1 was also found to enhance p21 transcription by inhibiting Wnt/β-catenin signaling. We identified histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2, as a target of Wnt/β-catenin signaling. HBP1-mediated repression of EZH2 through Wnt/β-catenin signaling decreased the level of trimethylation of histone H3 at lysine 27 of overall and specific histone on the p21 promoter, resulting in p21 transactivation. Although intricate, the reciprocal partnership of HBP1 and p21 has exceptional importance. HBP1-mediated elevation of p21 through the Mdm2/p53 and TCF4/EZH2 pathways contributes to both cellular senescence and tumor inhibition. Together, our results suggest that the HBP1 transcription factor orchestrates a complex regulation of key genes during cellular senescence and tumorigenesis with an impact on protein ubiquitination and overall histone methylation state.
Collapse
Affiliation(s)
- Yifan Chen
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Kewu Pan
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Pingzhang Wang
- the Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhengyi Cao
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Weibin Wang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Shuya Wang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Ningguang Hu
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Junhui Xue
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Hui Li
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Wei Jiang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Gang Li
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and
| | - Xiaowei Zhang
- From the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191 and.
| |
Collapse
|
39
|
Thorley M, Malatras A, Duddy W, Le Gall L, Mouly V, Butler Browne G, Duguez S. Changes in Communication between Muscle Stem Cells and their Environment with Aging. J Neuromuscul Dis 2015; 2:205-217. [PMID: 27858742 PMCID: PMC5240546 DOI: 10.3233/jnd-150097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging is associated with both muscle weakness and a loss of muscle mass, contributing towards overall frailty in the elderly. Aging skeletal muscle is also characterised by a decreasing efficiency in repair and regeneration, together with a decline in the number of adult stem cells. Commensurate with this are general changes in whole body endocrine signalling, in local muscle secretory environment, as well as in intrinsic properties of the stem cells themselves. The present review discusses the various mechanisms that may be implicated in these age-associated changes, focusing on aspects of cell-cell communication and long-distance signalling factors, such as levels of circulating growth hormone, IL-6, IGF1, sex hormones, and inflammatory cytokines. Changes in the local environment are also discussed, implicating IL-6, IL-4, FGF-2, as well as other myokines, and processes that lead to thickening of the extra-cellular matrix. These factors, involved primarily in communication, can also modulate the intrinsic properties of muscle stem cells, including reduced DNA accessibility and repression of specific genes by methylation. Finally we discuss the decrease in the stem cell pool, particularly the failure of elderly myoblasts to re-quiesce after activation, and the consequences of all these changes on general muscle homeostasis.
Collapse
Affiliation(s)
- Matthew Thorley
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Apostolos Malatras
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - William Duddy
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Laura Le Gall
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Gillian Butler Browne
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Stéphanie Duguez
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| |
Collapse
|
40
|
Zhang M, Xu W, Ke M, Xu J, Deng Y. Evaluation of individual aging degree by standard-free, label-free LC-MS/MS quantification of formaldehyde-modified peptides. Analyst 2015; 140:4137-42. [PMID: 25905659 DOI: 10.1039/c5an00355e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a standard-free, label-free LC-MS/MS method is proposed to evaluate aging based on the cross-linkage theory. First, an aging-biomarker screening model was set up in vitro with formaldehyde and the most abundant protein in plasma, human serum albumin (HSA), based on the Maillard reaction. The modification level of peptides cleaved from HSA was investigated using a liquid chromatography tandem mass spectrometry (LC-MS/MS) method with an (18)O-labeling technique. One formaldehyde-insensitive peptide and six formaldehyde-sensitive peptides that would be verified for being putative peptide-biomarkers were screened via the in vitro aging model. These six putative biomarkers were then preliminarily verified by plasma samples with the aldehyde-insensitive peptide serving as the internal standard. The verification results indicated that the peptides LDELRDEGK and VFDEFKPLVEEPQNLIK showed a significant quantitative difference among young/mid-aged/elderly groups of people.
Collapse
Affiliation(s)
- Mei Zhang
- National Institute for Communicable Disease Control and Prevention, Beijing, 102206, China.
| | | | | | | | | |
Collapse
|
41
|
Yablonka-Reuveni Z, Danoviz ME, Phelps M, Stuelsatz P. Myogenic-specific ablation of Fgfr1 impairs FGF2-mediated proliferation of satellite cells at the myofiber niche but does not abolish the capacity for muscle regeneration. Front Aging Neurosci 2015; 7:85. [PMID: 26074812 PMCID: PMC4446549 DOI: 10.3389/fnagi.2015.00085] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 04/30/2015] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle satellite cells (SCs) are Pax7+ myogenic stem cells that reside between the basal lamina and the plasmalemma of the myofiber. In mature muscles, SCs are typically quiescent, but can be activated in response to muscle injury. Depending on the magnitude of tissue trauma, SCs may divide minimally to repair subtle damage within individual myofibers or produce a larger progeny pool that forms new myofibers in cases of overt muscle injury. SC transition through proliferation, differentiation and renewal is governed by the molecular blueprint of the cells as well as by the extracellular milieu at the SC niche. In particular, the role of the fibroblast growth factor (FGF) family in regulating SCs during growth and aging is well recognized. Of the several FGFs shown to affect SCs, FGF1, FGF2, and FGF6 proteins have been documented in adult skeletal muscle. These prototypic paracrine FGFs transmit their mitogenic effect through the FGFRs, which are transmembrane tyrosine kinase receptors. Using the mouse model, we show here that of the four Fgfr genes, only Fgfr1 and Fgfr4 are expressed at relatively high levels in quiescent SCs and their proliferating progeny. To further investigate the role of FGFR1 in adult myogenesis, we have employed a genetic (Cre/loxP) approach for myogenic-specific (MyoDCre-driven) ablation of Fgfr1. Neither muscle histology nor muscle regeneration following cardiotoxin-induced injury were overtly affected in Fgfr1-ablated mice. This suggests that FGFR1 is not obligatory for SC performance in this acute muscle trauma model, where compensatory growth factor/cytokine regulatory cascades may exist. However, the SC mitogenic response to FGF2 is drastically repressed in isolated myofibers prepared from Fgfr1-ablated mice. Collectively, our study indicates that FGFR1 is important for FGF-mediated proliferation of SCs and its mitogenic role is not compensated by FGFR4 that is also highly expressed in SCs.
Collapse
Affiliation(s)
- Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Maria E Danoviz
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Michael Phelps
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Pascal Stuelsatz
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| |
Collapse
|
42
|
Skeletal muscle microRNA and messenger RNA profiling in cofilin-2 deficient mice reveals cell cycle dysregulation hindering muscle regeneration. PLoS One 2015; 10:e0123829. [PMID: 25874796 PMCID: PMC4395318 DOI: 10.1371/journal.pone.0123829] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 03/07/2015] [Indexed: 12/19/2022] Open
Abstract
Congenital myopathies are rare skeletal muscle diseases presenting in early age with hypotonia and weakness often linked to a genetic defect. Mutations in the gene for cofilin-2 (CFL2) have been identified in several families as a cause of congenital myopathy with nemaline bodies and cores. Here we explore the global messenger and microRNA expression patterns in quadriceps muscle samples from cofillin-2-null mice and compare them with sibling-matched wild-type mice to determine the molecular pathways and mechanisms involved. Cell cycle processes are markedly dysregulated, with altered expression of genes involved in mitotic spindle formation, and evidence of loss of cell cycle checkpoint regulation. Importantly, alterations in cell cycle, apoptosis and proliferation pathways are present in both mRNA and miRNA expression patterns. Specifically, p21 transcript levels were increased, and the expression of p21 targets, such as cyclin D and cyclin E, was decreased. We therefore hypothesize that deficiency of cofilin-2 is associated with interruption of the cell cycle at several checkpoints, hindering muscle regeneration. Identification of these pathways is an important step towards developing appropriate therapies against various congenital myopathies.
Collapse
|