1
|
Wang Z, Peng Q, Zhang Z, You X, Duan H, Sha R, Yuan N, Li Z, Xie Z, Han J, Feng Y. SRSF1 Is Crucial for Maintaining Satellite Cell Homeostasis During Skeletal Muscle Growth and Regeneration. J Cachexia Sarcopenia Muscle 2024; 15:2629-2641. [PMID: 39381961 PMCID: PMC11634495 DOI: 10.1002/jcsm.13607] [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: 12/13/2023] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The splicing factor SRSF1 emerges as a mater regulator of cell proliferation, displaying high expression in actively proliferative satellite cells (SCs). In SRSF1 knockout mice (KO) generated via MyoD-Cre, early mortality and muscle atrophy are observed during postnatal muscle growth. Despite these findings, the precise mechanisms through which SRSF1 loss influences SCs' functions and its role in muscle regeneration remain to be elucidated. METHODS To unravel the exact mechanisms underlying the impact of SRSF1 deficiency SC functions, we employed single-cell RNA sequencing (scRNA-seq) on a mononuclear cell suspension isolated from the newborn diaphragm of KO and control mice. Concurrently, we subjected diaphragm muscles to RNA-seq analysis to identify dysregulated splicing events associated with SRSF1 deletion. For the analysis of the effect of SRSF1 deletion on muscle regeneration, we generated mice with inducible SC-specific Srsf1 ablation through Pax7-CreER. SRSF1 ablation was induced by intraperitoneal injection of tamoxifen. Using cardiotoxin-induced muscle injury, we examined the consequences of SRSF1 depletion on SC function through HE staining, immunostaining and EdU incorporation assay. C2C12 myoblasts and isolated myoblasts were employed to assess stem cell function and senescence. RESULTS Utilizing scRNA-seq analysis, we observed a noteworthy increase in activated and proliferating myoblasts when SRSF1 was absent. This increase was substantial, with the proportion rising from 28.68% in the control group to 77.06% in the knockout group. However, these myoblasts experienced mitotic abnormalities in the absence of SRSF1, resulting in cell cycle arrest and the onset of cellular senescence. In the knockout mice, the proportion of Pax7+ cells within improper niche positioning increased significantly to 25% compared to 12% in the control cells (n ≥ 10, p < 0.001). Furthermore, there was an observation of persistent cell cycle exit specifically in the Pax7+ cells deficient in SRSF1 (n = 6, p < 0.001). SRSF1 plays a pivotal role in regulating the splicing of Fgfr1op2, favouring the full-length isoform crucial for mitotic spindle organization. Disrupting SRSF1 in C2C12 and primary myoblasts results in multipolar spindle formation (p < 0.001) and dysregulated splicing of Fgfr1op2 and triggers cellular senescence. Consequently, adult SCs lacking SRSF1 initially activate upon injury but face substantial challenge in proliferation (n = 4, p < 0.001), leading to a failure in muscle regeneration. CONCLUSIONS SRSF1 plays a critical role in SCs by ensuring proper splicing, maintaining mitotic progression and preventing premature senescence. These findings underscore the significant role of SRSF1 in controlling SC proliferation during skeletal muscle growth and regeneration.
Collapse
Affiliation(s)
- Zhenzhen Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Qian Peng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Zhige Zhang
- Department of General Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xue You
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical UniversityJiningChina
| | - Huimin Duan
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical UniversityJiningChina
| | - Rula Sha
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Ningyang Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Zhigang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Zhiqin Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Jun Han
- Department of General Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Ying Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical UniversityJiningChina
| |
Collapse
|
2
|
Yuan G, Yu C, Du X, Li D, Dou H, Lu P, Wu T, Hao C, Wang Y. Injectable GelMA Hydrogel Microspheres with Sustained Release of Platelet-Rich Plasma for the Treatment of Thin Endometrium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403890. [PMID: 39206600 DOI: 10.1002/smll.202403890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Platelet-rich plasma (PRP) intrauterine infusion has been demonstrated to be effective in treating thin endometrium and achieving pregnancy. However, the rapid release of growth factors limits its effectiveness in clinical applications, and thus, multiple intrauterine infusions are often required to achieve therapeutic efficacy. In this study, a GelMA hydrogel microsphere biomaterial is developed using droplet microfluidics to modify the delivery mode of PRP and thus prolong its duration of action. Its biocompatibility is confirmed through both in vivo and in vitro studies. Cell experiments show that PRP-loaded microspheres significantly enhance cell proliferation, migration, and angiogenesis. In vivo experiments show that the effects of PRP-loaded microspheres on repairing the endometrium and restoring fertility in mice could achieve the impact of triple PRP intrauterine infusions. Further mechanistic investigations reveal that PRP could facilitate endometrial repair by regulating the expression of E2Fs, a group of transcription factors. This study demonstrates that hydrogel microspheres could modify the delivery of PRP and prolong its duration of action, enabling endometrial repair and functional reconstruction. This design avoids repeated intrauterine injections of PRP in the clinic, reduces the number of patient visits, and provides a new avenue for clinical treatment of thin endometrium.
Collapse
Affiliation(s)
- Guanghui Yuan
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Chenghao Yu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China
| | - Xin Du
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Duan Li
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Huaiqian Dou
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Panpan Lu
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Tong Wu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Qingdao University, Qingdao, 266071, China
| | - Cuifang Hao
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| |
Collapse
|
3
|
Lagunas-Rangel FA, Liepinsh E, Fredriksson R, Alsehli AM, Williams MJ, Dambrova M, Jönsson J, Schiöth HB. Off-target effects of statins: molecular mechanisms, side effects and the emerging role of kinases. Br J Pharmacol 2024; 181:3799-3818. [PMID: 39180421 DOI: 10.1111/bph.17309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 08/26/2024] Open
Abstract
Statins are one of the most important classes of drugs. In this analytical review, we elucidate the intricate molecular mechanisms and toxicological rationale regarding both the on- (targeting 3-hydroxy-3-methylglutaryl-coenzyme A reductase [HMGCR]) and off-target effects of statins. Statins interact with a number of membrane kinases, such as epidermal growth factor receptor (EGFR), erb-b2 receptor tyrosine kinase 2 (HER2) and MET proto-oncogene, receptor tyrosine kinase (MET), as well as cytosolic kinases, such as SRC proto-oncogene, non-receptor tyrosine kinase (Src) and show inhibitory activity at nanomolar concentrations. In addition, they interact with calcium ATPases and peroxisome proliferator-activated receptor α (PPARα/NR1C1) at higher concentrations. Statins interact with mitochondrial complexes III and IV, and their inhibition of coenzyme Q10 synthesis also impairs the functioning of complexes I and II. Statins act as inhibitors of kinases, calcium ATPases and mitochondrial complexes, while activating PPARα. These off-target effects likely contribute to the side effects observed in patients undergoing statin therapy, including musculoskeletal symptoms and hepatic effects. Interestingly, some off-target effects of statins could also be the cause of favourable outcomes, relating to repurposing statins in conditions such as inflammatory disorders and cancer.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Edgars Liepinsh
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ahmed M Alsehli
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Michael J Williams
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Riga Stradiņš University, Riga, Latvia
| | - Jörgen Jönsson
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Sha R, Guo R, Duan H, Peng Q, Yuan N, Wang Z, Li Z, Xie Z, You X, Feng Y. SRSF2 is a key player in orchestrating the directional migration and differentiation of MyoD progenitors during skeletal muscle development. eLife 2024; 13:RP98175. [PMID: 39248331 PMCID: PMC11383525 DOI: 10.7554/elife.98175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Abstract
SRSF2 plays a dual role, functioning both as a transcriptional regulator and a key player in alternative splicing. The absence of Srsf2 in MyoD + progenitors resulted in perinatal mortality in mice, accompanied by severe skeletal muscle defects. SRSF2 deficiency disrupts the directional migration of MyoD progenitors, causing them to disperse into both muscle and non-muscle regions. Single-cell RNA-sequencing analysis revealed significant alterations in Srsf2-deficient myoblasts, including a reduction in extracellular matrix components, diminished expression of genes involved in ameboid-type cell migration and cytoskeleton organization, mitosis irregularities, and premature differentiation. Notably, one of the targets regulated by Srsf2 is the serine/threonine kinase Aurka. Knockdown of Aurka led to reduced cell proliferation, disrupted cytoskeleton, and impaired differentiation, reflecting the effects seen with Srsf2 knockdown. Crucially, the introduction of exogenous Aurka in Srsf2-knockdown cells markedly alleviated the differentiation defects caused by Srsf2 knockdown. Furthermore, our research unveiled the role of Srsf2 in controlling alternative splicing within genes associated with human skeletal muscle diseases, such as BIN1, DMPK, FHL1, and LDB3. Specifically, the precise knockdown of the Bin1 exon17-containing variant, which is excluded following Srsf2 depletion, profoundly disrupted C2C12 cell differentiation. In summary, our study offers valuable insights into the role of SRSF2 in governing MyoD progenitors to specific muscle regions, thereby controlling their differentiation through the regulation of targeted genes and alternative splicing during skeletal muscle development.
Collapse
Affiliation(s)
- Rula Sha
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruochen Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huimin Duan
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical University, Jining, China
| | - Qian Peng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ningyang Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenzhen Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhigang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiqin Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xue You
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical University, Jining, China
| | - Ying Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
5
|
Folgueira C, Herrera-Melle L, López JA, Galvan-Alvarez V, Martin-Rincon M, Cuartero MI, García-Culebras A, Dumesic PA, Rodríguez E, Leiva-Vega L, León M, Porteiro B, Iglesias C, Torres JL, Hernández-Cosido L, Bonacasa C, Marcos M, Moro MÁ, Vázquez J, Calbet JAL, Spiegelman BM, Mora A, Sabio G. Remodeling p38 signaling in muscle controls locomotor activity via IL-15. SCIENCE ADVANCES 2024; 10:eadn5993. [PMID: 39141732 PMCID: PMC11323882 DOI: 10.1126/sciadv.adn5993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/10/2024] [Indexed: 08/16/2024]
Abstract
Skeletal muscle has gained recognition as an endocrine organ releasing myokines upon contraction during physical exercise. These myokines exert both local and pleiotropic health benefits, underscoring the crucial role of muscle function in countering obesity and contributing to the overall positive effects of exercise on health. Here, we found that exercise activates muscle p38γ, increasing locomotor activity through the secretion of interleukin-15 (IL-15). IL-15 signals in the motor cortex, stimulating locomotor activity. This activation of muscle p38γ, leading to an increase locomotor activity, plays a crucial role in reducing the risk of diabetes and liver steatosis, unveiling a vital muscle-brain communication pathway with profound clinical implications. The correlation between p38γ activation in human muscle during acute exercise and increased blood IL-15 levels highlights the potential therapeutic relevance of this pathway in treating obesity and metabolic diseases. These findings provide valuable insights into the molecular basis of exercise-induced myokine responses promoting physical activity.
Collapse
Affiliation(s)
- Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Juan Antonio López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Victor Galvan-Alvarez
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria 35017, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Marcos Martin-Rincon
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria 35017, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Canary Islands, Spain
| | - María Isabel Cuartero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Alicia García-Culebras
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Phillip A. Dumesic
- Dana Farber Cancer Institute (DFCI), Department of Cell Biology, Harvard University Medical School, Boston, MA, USA
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Marta León
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Begoña Porteiro
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Cristina Iglesias
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | | | - Lourdes Hernández-Cosido
- Bariatric Surgery Unit, Department of General Surgery, University Hospital of Salamanca, Department of Surgery, University of Salamanca, Salamanca, Spain
| | - Clara Bonacasa
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Salamanca, Spain; Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María Ángeles Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jose A. L. Calbet
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria 35017, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Canary Islands, Spain
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Bruce M. Spiegelman
- Dana Farber Cancer Institute (DFCI), Department of Cell Biology, Harvard University Medical School, Boston, MA, USA
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
6
|
Otsuka T, Kan HM, Mengsteab PY, Tyson B, Laurencin CT. Fibroblast growth factor 8b (FGF-8b) enhances myogenesis and inhibits adipogenesis in rotator cuff muscle cell populations in vitro. Proc Natl Acad Sci U S A 2024; 121:e2314585121. [PMID: 38147545 PMCID: PMC10769839 DOI: 10.1073/pnas.2314585121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/24/2023] [Indexed: 12/28/2023] Open
Abstract
Fatty expansion is one of the features of muscle degeneration due to muscle injuries, and its presence interferes with muscle regeneration. Specifically, poor clinical outcomes have been linked to fatty expansion in rotator cuff tears and repairs. Our group recently found that fibroblast growth factor 8b (FGF-8b) inhibits adipogenic differentiation and promotes myofiber formation of mesenchymal stem cells in vitro. This led us to hypothesize that FGF-8b could similarly control the fate of muscle-specific cell populations derived from rotator cuff muscle involved in muscle repair following rotator cuff injury. In this study, we isolate fibro-adipogenic progenitor cells (FAPs) and satellite stem cells (SCs) from rat rotator cuff muscle tissue and analyzed the effects of FGF-8b supplementation. Utilizing a cell plating protocol, we successfully isolate FAPs-rich fibroblasts (FIBs) and SCs-rich muscle progenitor cells (MPCs). Subsequently, we demonstrate that FIB adipogenic differentiation can be inhibited by FGF-8b, while MPC myogenic differentiation can be enhanced by FGF-8b. We further demonstrate that phosphorylated ERK due to FGF-8b leads to the inhibition of adipogenesis in FIBs and SCs maintenance and myofiber formation in MPCs. Together, these findings demonstrate the powerful potential of FGF-8b for rotator cuff repair by altering the fate of muscle undergoing degeneration.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences, University of Connecticut Health Center, Farmington, CT06030
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences, University of Connecticut Health Center, Farmington, CT06030
| | - Paulos Y. Mengsteab
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences, University of Connecticut Health Center, Farmington, CT06030
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT06030
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT06269
| | - Breajah Tyson
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences, University of Connecticut Health Center, Farmington, CT06030
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT06030
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT06269
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT06269
| |
Collapse
|