1
|
L C, P M, I B, A R, H D, D S. Obesity-driven musculotendinous remodeling impairs tissue resilience to mechanical damage. Cell Tissue Res 2025:10.1007/s00441-025-03967-1. [PMID: 40163175 DOI: 10.1007/s00441-025-03967-1] [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: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Obesity has been associated with lower muscle strength-to-body mass ratio. Here, we evaluated the effects of diet-induced obesity on the mechano-structural properties of isolated muscles and tendons. Thirty 10-week-old male C57BL/6 J mice were randomly assigned to either an obesogenic high-fat diet group (OB) for 24 weeks or a control group (CN) maintained on a standard chow diet. Soleus muscle (SOL) and Achilles tendon (AT) specimens were isolated and subjected either to failure testing, 300 cycles of passive stretch-destretch, or isometric twitch contractions. Morpho-structural and protein expression analyses were conducted to assess collagen and adipose tissue accumulation, concentrations of cross-linking factors, and any alterations in the POSTN-TGFβ1-Akt signaling pathway. OB SOL and AT tissues were more fragile than those from CN (p < 0.05). A piecewise linear regression model revealed a tendency for OB tissues to exhibit steeper mechanical property changes within the first 20 cycles compared to CN, followed by a similar plateau phase in both groups. OB SOL-AT complexes showed a slower twitch-contraction-relaxation pattern than CN (p < 0.05). OB tendons and muscles were larger than those of the CN, with muscles featuring bigger fibers, and higher collagen area fraction (p < 0.05). Elevated TGFβ1 and POSTN concentrations were observed in OB tissues (p < 0.05), alongside increased P-Akt and P-4EBP1 expression (p < 0.05). These findings highlight the detrimental effects of obesity on the structural integrity of muscle and tendon tissues and suggest a significant role of POSTN-TGFβ1-Akt signaling in obesity-associated musculotendinous remodeling.
Collapse
Affiliation(s)
- Cesanelli L
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania.
| | - Minderis P
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Balnyte I
- Department of Histology and Embryology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ratkevicius A
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
- Sports and Exercise Medicine Centre, Queen Mary University of London, London, UK
| | - Degens H
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Satkunskiene D
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
2
|
Hurley-Novatny A, Chang D, Murakami K, Wang L, Li H. Poor bone health in Duchenne muscular dystrophy: a multifactorial problem beyond corticosteroids and loss of ambulation. Front Endocrinol (Lausanne) 2024; 15:1398050. [PMID: 39669499 PMCID: PMC11634624 DOI: 10.3389/fendo.2024.1398050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, fatal muscle wasting disease caused by X-linked mutations in the dystrophin gene. Alongside the characteristic muscle weakness, patients face a myriad of skeletal complications, including osteoporosis/osteopenia, high susceptibility to vertebral and long bone fractures, fat embolism post-fracture, scoliosis, and growth retardation. Those skeletal abnormalities significantly compromise quality of life and are sometimes life-threatening. These issues were traditionally attributed to loss of ambulation and chronic corticosteroid use, but recent investigations have unveiled a more intricate etiology. Factors such as vitamin D deficiency, hormonal imbalances, systemic inflammation, myokine release from dystrophic muscle, and vascular dysfunction are emerging as significant contributors as well. This expanded understanding illuminates the multifaceted pathogenesis underlying skeletal issues in DMD. Present therapeutic options are limited and lack specificity. Advancements in understanding the pathophysiology of bone complications in DMD will offer promising avenues for novel treatment modalities. In this review, we summarize the current understanding of factors contributing to bone problems in DMD and delineate contemporary and prospective multidisciplinary therapeutic approaches.
Collapse
Affiliation(s)
- Amelia Hurley-Novatny
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - David Chang
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Katsuhiro Murakami
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Ling Wang
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Hongshuai Li
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
3
|
Yokomizo-Goto M, Takenaka-Ninagawa N, Zhao C, Bourgeois Yoshioka CK, Miki M, Motoike S, Inada Y, Zujur D, Theoputra W, Jin Y, Toguchida J, Ikeya M, Sakurai H. Distinct muscle regenerative capacity of human induced pluripotent stem cell-derived mesenchymal stromal cells in Ullrich congenital muscular dystrophy model mice. Stem Cell Res Ther 2024; 15:340. [PMID: 39370505 PMCID: PMC11457425 DOI: 10.1186/s13287-024-03951-6] [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: 07/17/2024] [Accepted: 09/18/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Ullrich congenital muscular dystrophy (UCMD) is caused by a deficiency in type 6 collagen (COL6) due to mutations in COL6A1, COL6A2, or COL6A3. COL6 deficiency alters the extracellular matrix structure and biomechanical properties, leading to mitochondrial defects and impaired muscle regeneration. Therefore, mesenchymal stromal cells (MSCs) that secrete COL6 have attracted attention as potential therapeutic targets. Various tissue-derived MSCs exert therapeutic effects in various diseases. However, no reports have compared the effects of MSCs of different origins on UCMD pathology. METHODS To evaluate which MSC population has the highest therapeutic efficacy for UCMD, in vivo (transplantation of MSCs to Col6a1-KO/NSG mice) and in vitro experiments (muscle stem cell [MuSCs] co-culture with MSCs) were conducted using adipose tissue-derived MSCs, bone marrow-derived MSCs, and xeno-free-induced iPSC-derived MSCs (XF-iMSCs). RESULTS In transplantation experiments on Col6a1-KO/NSG mice, the group transplanted with XF-iMSCs showed significantly enhanced muscle fiber regeneration compared to the other groups 1 week after transplantation. At 12 weeks after transplantation, only the XF-iMSCs transplantation group showed a significantly larger muscle fiber diameter than the other groups without inducing fibrosis, which was observed in the other transplantation groups. Similarly, in co-culture experiments, XF-iMSCs were found to more effectively promote the fusion and differentiation of MuSCs derived from Col6a1-KO/NSG mice than the other primary MSCs investigated in this study. Additionally, in vitro knockdown and supplementation experiments suggested that the IGF2 secreted by XF-iMSCs promoted MuSC differentiation. CONCLUSION XF-iMSCs are promising candidates for promoting muscle regeneration while avoiding fibrosis, offering a safer and more effective therapeutic approach for UCMD than other potential therapies.
Collapse
Affiliation(s)
- Megumi Yokomizo-Goto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Nana Takenaka-Ninagawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan.
| | - Chengzhu Zhao
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Clémence Kiho Bourgeois Yoshioka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Mayuho Miki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Souta Motoike
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yoshiko Inada
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Denise Zujur
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - William Theoputra
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yonghui Jin
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Junya Toguchida
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
4
|
Nakata K, Ishikawa M, Kamei N, Miyaki S, Adachi N, Inoue K, Kawabata S. Skeletal muscle injury treatment using the Silk Elastin® injection in a rat model. Regen Ther 2024; 26:180-187. [PMID: 38948131 PMCID: PMC11214263 DOI: 10.1016/j.reth.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/04/2024] [Accepted: 05/19/2024] [Indexed: 07/02/2024] Open
Abstract
Background Skeletal muscle injury (SMI) is often treated conservatively, although it can lead to scar tissue formation, which impedes muscle function and increases muscle re-injury risk. However, effective interventions for SMIs are yet to be established. Hypothesis The administration of Silk Elastin® (SE), a novel artificial protein, to the SMI site can suppress scar formation and promote tissue repair. Study design A controlled laboratory study. Methods In vitro: Fibroblast migration ability was assessed using a scratch assay. SE solution was added to the culture medium, and the fibroblast migration ability was compared across different concentrations. In vivo: An SMI model was established with Sprague-Dawley rats, which were assigned to three groups based on the material injected to the SMI site: SE gel (SE group; n = 8), atelocollagen gel (Atelo group; n = 8), and phosphate buffer saline (PBS group; n = 8). Histological evaluations were performed at weeks 1 and 4 following the SMI induction. In the 1-week model, we detected the expression of transforming growth factor (TGF)-β1 in the stroma using immunohistological evaluation and real-time polymerase chain reaction analysis. In the 4-week model, we measured tibialis anterior muscle strength upon peroneal nerve stimulation as a functional assessment. Results In vitro: The fibroblast migration ability was suppressed by SE added at a concentration of 10⁴ μg/mL in the culture medium. In vivo: In the 1-week model, the SE group exhibited significantly lower TGFβ -1 expression than the PBS group. In the 4-week model, the SE group had a significantly larger regenerated muscle fiber diameter and smaller scar formation area ratio than the other two groups. Moreover, the SE group was superior to the other two groups in terms of regenerative muscle strength. Conclusion Injection of SE gel to the SMI site may inhibit tissue scarring by reducing excessive fibroblast migration, thereby enhancing tissue repair. Clinical relevance The findings of this study may contribute to the development of an early intervention method for SMIs.
Collapse
Affiliation(s)
- Kyohei Nakata
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Japan
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Faculty of Medicine, Kagawa University Hospital, Japan
| | - Naosuke Kamei
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Japan
| | - Shigeru Miyaki
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Japan
| | | | | |
Collapse
|
5
|
Na Nan D, Klincumhom N, Trachoo V, Everts V, Ferreira JN, Osathanon T, Pavasant P. Periostin-integrin interaction regulates force-induced TGF-β1 and α-SMA expression by hPDLSCs. Oral Dis 2024; 30:2570-2579. [PMID: 37466141 DOI: 10.1111/odi.14691] [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: 01/07/2023] [Revised: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVE Periostin (PN), a major matricellular periodontal ligament (PDL) protein, modulates the remodeling of the PDL and bone, especially under mechanical stress. This study investigated the requirement of PN-integrin signaling in force-induced expression of transforming growth factor-beta 1 (TGF-β1) and alpha-smooth muscle actin (α-SMA) in human PDL stem cells (hPDLSCs). METHODS Cells were stimulated with intermittent compressive force (ICF) using computerized controlled apparatus. Cell migration was examined using in vitro scratch assay. The mRNA expression was examined using real-time polymerase chain reaction. The protein expression was determined using immunofluorescent staining and western blot analysis. RESULTS Stimulation with ICF for 24 h increased the expression of PN, TGF-β1, and α-SMA, along with increased SMAD2/3 phosphorylation. Knockdown of POSTN (PN gene) decreased the protein levels of TGF-β1 and pSMAD2/3 upon force stimulation. POSTN knockdown of hPDLSCs resulted in delayed cell migration, as determined by a scratch assay. However, migration improved after seeding these knockdown cells on pre-PN-coated surfaces. Further, the knockdown of αVβ5 significantly attenuated the force-induced TGF-β1 expression. CONCLUSION Our findings indicate the importance of PN-αVβ5 interactions in ICF-induced TGF-β1 signaling and the expression of α-SMA. Findings support the critical role of PN in maintaining the PDL's tissue integrity and homeostasis.
Collapse
Affiliation(s)
- Daneeya Na Nan
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nuttha Klincumhom
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vincent Everts
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Oral Cell Biology, Faculty of Dentistry, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Joao N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Qiao B, Liu X, Wang B, Wei S. The role of periostin in cardiac fibrosis. Heart Fail Rev 2024; 29:191-206. [PMID: 37870704 DOI: 10.1007/s10741-023-10361-y] [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] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Cardiac fibrosis, which is the buildup of proteins in the connective tissues of the heart, can lead to end-stage extracellular matrix (ECM) remodeling and ultimately heart failure. Cardiac remodeling involves changes in gene expression in cardiac cells and ECM, which significantly leads to the morbidity and mortality in heart failure. However, despite extensive research, the elusive intricacies underlying cardiac fibrosis remain unidentified. Periostin, an extracellular matrix (ECM) protein of the fasciclin superfamily, acts as a scaffold for building complex architectures in the ECM, which improves intermolecular interactions and augments the mechanical properties of connective tissues. Recent research has shown that periostin not only contributes to normal ECM homeostasis in a healthy heart but also serves as a potent inducible regulator of cellular reorganization in cardiac fibrosis. Here, we reviewed the constitutive domain of periostin and its interaction with other ECM proteins. We have also discussed the critical pathophysiological functions of periostin in cardiac remodeling mechanisms, including two distinct yet potentially intertwined mechanisms. Furthermore, we will focus on the intrinsic complexities within periostin research, particularly surrounding the contentious issues observed in experimental findings.
Collapse
Affiliation(s)
- Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
7
|
Xu D, Wan B, Qiu K, Wang Y, Zhang X, Jiao N, Yan E, Wu J, Yu R, Gao S, Du M, Liu C, Li M, Fan G, Yin J. Single-Cell RNA-Sequencing Provides Insight into Skeletal Muscle Evolution during the Selection of Muscle Characteristics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305080. [PMID: 37870215 PMCID: PMC10724408 DOI: 10.1002/advs.202305080] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/27/2023] [Indexed: 10/24/2023]
Abstract
Skeletal muscle comprises a large, heterogeneous assortment of cell populations that interact to maintain muscle homeostasis, but little is known about the mechanism that controls myogenic development in response to artificial selection. Different pig (Sus scrofa) breeds exhibit distinct muscle phenotypes resulting from domestication and selective breeding. Using unbiased single-cell transcriptomic sequencing analysis (scRNA-seq), the impact of artificial selection on cell profiles is investigated in neonatal skeletal muscle of pigs. This work provides panoramic muscle-resident cell profiles and identifies novel and breed-specific cells, mapping them on pseudotime trajectories. Artificial selection has elicited significant changes in muscle-resident cell profiles, while conserving signs of generational environmental challenges. These results suggest that fibro-adipogenic progenitors serve as a cellular interaction hub and that specific transcription factors identified here may serve as candidate target regulons for the pursuit of a specific muscle phenotype. Furthermore, a cross-species comparison of humans, mice, and pigs illustrates the conservation and divergence of mammalian muscle ontology. The findings of this study reveal shifts in cellular heterogeneity, novel cell subpopulations, and their interactions that may greatly facilitate the understanding of the mechanism underlying divergent muscle phenotypes arising from artificial selection.
Collapse
Affiliation(s)
- Doudou Xu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Boyang Wan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Kai Qiu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| | - Ning Jiao
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Enfa Yan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Jiangwei Wu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYangling712100China
| | - Run Yu
- Beijing National Day SchoolBeijing100039China
| | - Shuai Gao
- Key Laboratory of Animal GeneticsCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Min Du
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciences and School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
| | | | - Mingzhou Li
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengdu625014China
| | - Guoping Fan
- Department of Human GeneticsDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| |
Collapse
|
8
|
Yoshihara T, Morimoto T, Hirata H, Murayama M, Nonaka T, Tsukamoto M, Toda Y, Kobayashi T, Izuhara K, Mawatari M. Mechanisms of tissue degeneration mediated by periostin in spinal degenerative diseases and their implications for pathology and diagnosis: a review. Front Med (Lausanne) 2023; 10:1276900. [PMID: 38020106 PMCID: PMC10645150 DOI: 10.3389/fmed.2023.1276900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/18/2023] [Indexed: 12/01/2023] Open
Abstract
Periostin (POSTN) serves a dual role as both a matricellular protein and an extracellular matrix (ECM) protein and is widely expressed in various tissues and cells. As an ECM protein, POSTN binds to integrin receptors, transduces signals to cells, enabling cell activation. POSTN has been linked with various diseases, including atopic dermatitis, asthma, and the progression of multiple cancers. Recently, its association with orthopedic diseases, such as osteoporosis, osteoarthritis resulting from cartilage destruction, degenerative diseases of the intervertebral disks, and ligament degenerative diseases, has also become apparent. Furthermore, POSTN has been shown to be a valuable biomarker for understanding the pathophysiology of orthopedic diseases. In addition to serum POSTN, synovial fluid POSTN in joints has been reported to be useful as a biomarker. Risk factors for spinal degenerative diseases include aging, mechanical stress, trauma, genetic predisposition, obesity, and metabolic syndrome, but the cause of spinal degenerative diseases (SDDs) remains unclear. Studies on the pathophysiological effects of POSTN may significantly contribute toward the diagnosis and treatment of spinal degenerative diseases. Therefore, in this review, we aim to examine the mechanisms of tissue degeneration caused by mechanical and inflammatory stresses in the bones, cartilage, intervertebral disks, and ligaments, which are crucial components of the spine, with a focus on POSTN.
Collapse
Affiliation(s)
- Tomohito Yoshihara
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Tadatsugu Morimoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshihiro Nonaka
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masatsugu Tsukamoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Yu Toda
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takaomi Kobayashi
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
9
|
Mutsuzaki H, Kuwahara K, Nakajima H. Influence of periostin on the development of fibrocartilage layers of anterior cruciate ligament insertion. Orthop Traumatol Surg Res 2023; 109:103215. [PMID: 35092850 DOI: 10.1016/j.otsr.2022.103215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/11/2021] [Accepted: 07/16/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Periostin (Postn) is thought to play a role in the formation of anterior cruciate ligament (ACL) insertion. However, the influence of Postn on the development of ACL insertion requires further understanding. This study aimed to clarify the influence of Postn on the development of fibrocartilage layers of ACL insertion. HYPOTHESIS We hypothesized that Postn would influence the development of fibrocartilage layers of ACL insertion. MATERIALS AND METHODS C57BL/6N wild-type (Postn+/+; n=54) and Postn knockout (Postn-/-; n=54) mice were used in this study. Six animals were euthanized at 1 d and 1, 2, 3, 4, 6, 8, 10, and 12 weeks of age in each group. The chondrocyte number, proliferation, apoptosis, safranin O-stained glycosaminoglycan (GAG) area, type II collagen staining area, tidemark length, and insertion width were evaluated. RESULTS Chondrocyte proliferation was high up to 2 weeks in Postn+/+, while low at age 1 d; it was, especially lower in Postn-/- than in Postn+/+ at age 1 d and 1 week. Chondrocyte apoptosis was high up to age 8 weeks in Postn+/+ and at 6 weeks in Postn-/-; it was especially higher in Postn-/- than in Postn+/+ at age 1 week. The GAG stained area was thickest for age 1 d to 4 weeks in Postn+/+ and for age 2 to 6 weeks in Postn-/-. The type II collagen staining area in Postn+/+ was thicker than that in Postn-/- at age 6 and 8 weeks. The tidemark length in Postn+/+ was longer than that in Postn-/- from age 8 to 12 weeks. The insertion width in Postn+/+ was longer than that in Postn-/- from age 1 to 3 weeks. DISCUSSION Postn decreased cell proliferation in the early postnatal phase and influenced the development of the fibrocartilage layer extracellular matrix of ACL insertion in mice. Postn may contribute to the development of methods for regeneration of the ACL insertion. LEVEL OF EVIDENCE V; controlled laboratory study.
Collapse
Affiliation(s)
- Hirotaka Mutsuzaki
- Department of Orthopaedic Surgery, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami Ami-machi, Inashiki-gun, 300-0394 Ibaraki, Japan.
| | - Kazuki Kuwahara
- Department of Agriculture, Ibaraki University, 3-21-1 Chuo, Ami, 300-0393 Ibaraki, Japan
| | - Hiromi Nakajima
- Department of Agriculture, Ibaraki University, 3-21-1 Chuo, Ami, 300-0393 Ibaraki, Japan
| |
Collapse
|
10
|
Nguyen CDL, Jimenez-Moreno AC, Merker M, Bowers CJ, Nikolenko N, Hentschel A, Müntefering T, Isham A, Ruck T, Vorgerd M, Dobelmann V, Gourdon G, Schara-Schmidt U, Gangfuss A, Schröder C, Sickmann A, Gross C, Gorman G, Stenzel W, Kollipara L, Hathazi D, Spendiff S, Gagnon C, Preusse C, Duchesne E, Lochmüller H, Roos A. Periostin as a blood biomarker of muscle cell fibrosis, cardiomyopathy and disease severity in myotonic dystrophy type 1. J Neurol 2023; 270:3138-3158. [PMID: 36892629 DOI: 10.1007/s00415-023-11633-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 03/10/2023]
Abstract
BACKGROUND AND PURPOSE Myotonic dystrophy type 1 (DM1) is the most common form of adult-onset muscular dystrophy and is caused by an repeat expansion [r(CUG)exp] located in the 3' untranslated region of the DMPK gene. Symptoms include skeletal and cardiac muscle dysfunction and fibrosis. In DM1, there is a lack of established biomarkers in routine clinical practice. Thus, we aimed to identify a blood biomarker with relevance for DM1-pathophysiology and clinical presentation. METHODS We collected fibroblasts from 11, skeletal muscles from 27, and blood samples from 158 DM1 patients. Moreover, serum, cardiac, and skeletal muscle samples from DMSXL mice were included. We employed proteomics, immunostaining, qPCR and ELISA. Periostin level were correlated with CMRI-data available for some patients. RESULTS Our studies identified Periostin, a modulator of fibrosis, as a novel biomarker candidate for DM1: proteomic profiling of human fibroblasts and murine skeletal muscles showed significant dysregulation of Periostin. Immunostaining on skeletal and cardiac muscles from DM1 patients and DMSXL mice showed an extracellular increase of Periostin, indicating fibrosis. qPCR studies indicated increased POSTN expression in fibroblasts and muscle. Quantification of Periostin in blood samples from DMSXL mice and two large validation cohorts of DM1 patients showed decreased levels in animals and diseased individuals correlating with repeat expansion and disease severity and presence of cardiac symptoms identified by MRI. Analyses of longitudinal blood samples revealed no correlation with disease progression. CONCLUSIONS Periostin might serve as a novel stratification biomarker for DM1 correlating with disease severity, presence of cardiac malfunction and fibrosis.
Collapse
Affiliation(s)
- Chi D L Nguyen
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | | | - Monika Merker
- Department of Neurology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | | | | | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Thomas Müntefering
- Department of Neurology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Angus Isham
- Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Tobias Ruck
- Department of Neurology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Matthias Vorgerd
- Department of Neurology, University Hospital Bergmannsheil, Heimer Institute for Muscle Research, 44789, Bochum, Germany
| | - Vera Dobelmann
- Department of Neurology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Genevieve Gourdon
- Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- Laboratory CTGDM, Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Ulrike Schara-Schmidt
- Department of Neuropediatrics and Neuromuscular Centre for Children and Adolescents, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45147, Essen, Germany
| | - Andrea Gangfuss
- Department of Neuropediatrics and Neuromuscular Centre for Children and Adolescents, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45147, Essen, Germany
| | - Charlotte Schröder
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Claudia Gross
- Institute of Clinical Genetics and Tumor Genetics Bonn, Maximilianstraße 28D, 53111, Bonn, Germany
| | - Grainne Gorman
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227, Dortmund, Germany
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Sally Spendiff
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Cynthia Gagnon
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- School of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Corinna Preusse
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Elise Duchesne
- Department of Health Sciences, Université du Québec à Chicoutimi, Québec, Canada
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Centro Nacional de Análisis Genómico, Center for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Andreas Roos
- Department of Neurology, University Hospital Bergmannsheil, Heimer Institute for Muscle Research, 44789, Bochum, Germany.
- Department of Neuropediatrics and Neuromuscular Centre for Children and Adolescents, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45147, Essen, Germany.
- Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, The Ottawa Hospital, and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
11
|
Başımoğlu Koca Y, Koca S, Öztel Z, Balcan E. Determination of histopathological effects and myoglobin, periostin gene-protein expression levels in Danio rerio muscle tissue after acaricide yoksorrun-5EC (hexythiazox) application. Drug Chem Toxicol 2023; 46:50-58. [PMID: 34879781 DOI: 10.1080/01480545.2021.2007945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although pesticides are essential agrochemicals to annihilate harmful organisms in agriculture, their uncontrolled use has become an important threat to environmental health. Exposure to pesticides can affect many biological systems including immune system, endocrine system, and nervous system. However, the potential side effects of pesticides to skeletal muscle system remain unclear. Present study has focused on the evaluation of this issue by using an acaricide, yoksorrun-5EC (hexythiazox), in an aquatic model organism, Danio rerio. The histological analyses revealed that increased concentrations of the acaricide cause degradation of skeletal muscle along with increased necrosis and atrophy in myocytes, intercellular edema, and increased infiltrations between perimysium sheaths of muscle fibers. The effects of acaricide on myoglobin and periostin, which are associated with oxygen transport and muscle regeneration, respectively, were investigated at the gene and protein levels. RT-PCR results suggested that high concentration yoksorrun-5EC (hexythiazox) can induce myoglobin and periostin genes. Similar results were also obtained in the protein levels of these genes by western blotting analysis. These results suggested that yoksorrun-5EC (hexythiazox)-dependent disruption of skeletal muscle architecture is closely associated with the expression levels of myoglobin and periostin genes in Danio rerio model.
Collapse
Affiliation(s)
- Yücel Başımoğlu Koca
- Department of Biology, Zoology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Serdar Koca
- Department of Biology, General Biology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Zübeyde Öztel
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| | - Erdal Balcan
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
12
|
Hammers DW. NOX4 inhibition promotes the remodeling of dystrophic muscle. JCI Insight 2022; 7:158316. [PMID: 36278481 PMCID: PMC9714779 DOI: 10.1172/jci.insight.158316] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The muscular dystrophies (MDs) are genetic muscle diseases that result in progressive muscle degeneration followed by the fibrotic replacement of affected muscles as regenerative processes fail. Therapeutics that specifically address the fibrosis and failed regeneration associated with MDs represent a major unmet clinical need for MD patients, particularly those with advanced-stage disease progression. The current study investigated targeting NAD(P)H oxidase 4 (NOX4) as a potential strategy to reduce fibrosis and promote regeneration in disease-burdened muscle that models Duchenne muscular dystrophy (DMD). NOX4 was elevated in the muscles of dystrophic mice and DMD patients, localizing primarily to interstitial cells located between muscle fibers. Genetic and pharmacological targeting of NOX4 significantly reduced fibrosis in dystrophic respiratory and limb muscles. Mechanistically, NOX4 targeting decreased the number of fibrosis-depositing cells (myofibroblasts) and restored the number of muscle-specific stem cells (satellite cells) localized to their physiological niche, thereby rejuvenating muscle regeneration. Furthermore, acute inhibition of NOX4 was sufficient to induce apoptotic clearing of myofibroblasts within dystrophic muscle. These data indicate that targeting NOX4 is an effective strategy to promote the beneficial remodeling of disease-burdened muscle representative of DMD and, potentially, other MDs and muscle pathologies.
Collapse
Affiliation(s)
- David W. Hammers
- Department of Pharmacology & Therapeutics and
- Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
13
|
Mutsuzaki H, Yoshida Y, Nakajima H. Periostin Contributes to Fibrocartilage Layer Growth of the Patella Tendon Tibial Insertion in Mice. Medicina (B Aires) 2022; 58:medicina58070957. [PMID: 35888676 PMCID: PMC9319934 DOI: 10.3390/medicina58070957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: The influence of periostin on the growth of the patella tendon (PT) tibial insertion is unknown. The research described here aimed to reveal the contribution of periostin to the growth of fibrocartilage layers of the PT tibial insertion using periostin knockout mice. Materials and Methods: In both the wild-type (WD; C57BL/6N, periostin +/+; n = 54) and periostin knockout (KO; periostin −/−; n = 54) groups, six mice were euthanized on day 1 and at 1, 2, 3, 4, 6, 8, 10, and 12 weeks of age. Chondrocyte proliferation and apoptosis, number of chondrocytes, safranin O-stained glycosaminoglycan (GAG) area, staining area of type II collagen, and length of the tidemark were investigated. Results: Chondrocyte proliferation and apoptosis in KO were lower than those in WD on day 1 and at 1, 4, and 8 weeks and on day 1 and at 4, 6, and 12 weeks, respectively. Although the number of chondrocytes in both groups gradually decreased, it was lower in KO than in WD on day 1 and at 8 and 12 weeks. In the extracellular matrix, the GAG-stained area in KO was smaller than that in WD on day 1 and at 1, 4, 8, 10, and 12 weeks. The staining area of type II collagen in KO was smaller than that in WD at 8 weeks. The length of the tidemark in KO was shorter than that in WD at 4 and 6 weeks. Conclusion: Loss of periostin led to decreased chondrocyte proliferation, chondrocyte apoptosis, and the number of chondrocytes in the growth process of the PT tibial insertion. Moreover, periostin decreased and delayed GAG and type II collagen production and delayed tidemark formation in the growth process of the PT tibial insertion. Periostin can, therefore, contribute to the growth of fibrocartilage layers in the PT tibial insertion. Periostin deficiency may result in incomplete growth of the PT tibial insertion.
Collapse
Affiliation(s)
- Hirotaka Mutsuzaki
- Department of Orthopedic Surgery, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami Ami-machi, Inashiki-gun, Ibaraki, Ami 300-0394, Japan
- Correspondence: ; Tel.: +81-29-888-4000
| | - Yuta Yoshida
- Department of Agriculture, Ibaraki University, 3-21-1 Chuo, Ibaraki, Ami 300-0393, Japan; (Y.Y.); (H.N.)
| | - Hiromi Nakajima
- Department of Agriculture, Ibaraki University, 3-21-1 Chuo, Ibaraki, Ami 300-0393, Japan; (Y.Y.); (H.N.)
| |
Collapse
|
14
|
Zheng Y, Zeng L, Dong X, Du Q, Gao Y. Periostin aggravates the early phase of traumatic brain injury via the MAPK/ERK pathway. Neurol Res 2022; 44:560-569. [PMID: 35001858 DOI: 10.1080/01616412.2021.2024728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVES Periostin is found associated with trauma severity and mortality following head injury. In this study, the role and mechanism of periostin in the traumatic brain injury were investigated. METHODS Male Sprague-Dawley adult rats underwent sham or TBI modeling. Vehicle or recombinant periostin was administered intracerebroventricularly at 30 minutes post-TBI, and U0126, a specific MEK1/2 inhibitor, was administered intravenously at 30 minutes pre-TBI. Garcia neuroscore, limb function and brain water content assessments, as well as TUNEL and Western blotting assays were performed to evaluate the status of the above rats at 24 hours post-TBI. Finally, the motor test and Morris water maze test were performed to measure the effects of periostin and U0126 in the late phase of TBI. RESULTS Periostin expression significantly increased 24 hours post-TBI. Treatment with R-periostin aggravated post-TBI neurobehavioral impairment, brain edema, induced apoptosis and raised the quantity of phospho-p38, phospho-JNK, phospho-ERK and MMP-9, and lowered the expression of ZO-1. However, U0126, a kind of inhibitor of MEK, lowered the quantities of phospho-ERK and MMP-9, raised the expression of ZO-1, and suppressed apoptosis. U0126 also ameliorated the neurobehavioral impairments and brain edema induced by R-periostin. Additionally, U0126 didn't inhibit the expression of periostin in the early phase of TBI model. IU0126 was also able to ameliorate the pathological conditions in the late phase of TBI. DISCUSSION Periostin could aggravate neurobehavioral impairments and brain edema following TBI, and was involved in the early phase of TBI via the MAPK/ERK pathway.
Collapse
Affiliation(s)
- Yongke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Longhuan Zeng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yue Gao
- Department of Gerontology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Bus K, Szterk A. Relationship between Structure and Biological Activity of Various Vitamin K Forms. Foods 2021; 10:foods10123136. [PMID: 34945687 PMCID: PMC8701896 DOI: 10.3390/foods10123136] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/06/2023] Open
Abstract
Vitamin K is involved many biological processes, such as the regulation of blood coagulation, prevention of vascular calcification, bone metabolism and modulation of cell proliferation. Menaquinones (MK) and phylloquinone vary in biological activity, showing different bioavailability, half-life and transport mechanisms. Vitamin K1 and MK-4 remain present in the plasma for 8–24 h, whereas long-chain menaquinones can be detected up to 96 h after administration. Geometric structure is also an important factor that conditions their properties. Cis-phylloquinone shows nearly no biological activity. An equivalent study for menaquinone is not available. The effective dose to decrease uncarboxylated osteocalcin was six times lower for MK-7 than for MK-4. Similarly, MK-7 affected blood coagulation system at dose three to four times lower than vitamin K1. Both vitamin K1 and MK-7 inhibited the decline in bone mineral density, however benefits for the occurrence of cardiovascular diseases have been observed only for long-chain menaquinones. There are currently no guidelines for the recommended doses and forms of vitamin K in the prevention of osteoporosis, atherosclerosis and other cardiovascular disorders. Due to the presence of isomers with unknown biological properties in some dietary supplements, quality and safety of that products may be questioned.
Collapse
Affiliation(s)
- Katarzyna Bus
- Department of Spectrometric Methods, National Medicines Institute, 30/34 Chełmska, 00-725 Warsaw, Poland
- Correspondence:
| | - Arkadiusz Szterk
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797 Warsaw, Poland;
- Transfer of Science Sp. z o.o., Strzygłowska 15, 04-866 Warsaw, Poland
| |
Collapse
|
16
|
Shen X, Liu Z, Wang C, Xu F, Zhang J, Li M, Lei Y, Wang A, Bi C, Zhu G. Inhibition of Postn Rescues Myogenesis Defects in Myotonic Dystrophy Type 1 Myoblast Model. Front Cell Dev Biol 2021; 9:710112. [PMID: 34490258 PMCID: PMC8417118 DOI: 10.3389/fcell.2021.710112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/30/2021] [Indexed: 12/27/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an inherited neuromuscular disease caused by expanded CTG repeats in the 3' untranslated region (3'UTR) of the DMPK gene. The myogenesis process is defective in DM1, which is closely associated with progressive muscle weakness and wasting. Despite many proposed explanations for the myogenesis defects in DM1, the underlying mechanism and the involvement of the extracellular microenvironment remained unknown. Here, we constructed a DM1 myoblast cell model and reproduced the myogenesis defects. By RNA sequencing (RNA-seq), we discovered that periostin (Postn) was the most significantly upregulated gene in DM1 myogenesis compared with normal controls. This difference in Postn was confirmed by real-time quantitative PCR (RT-qPCR) and western blotting. Moreover, Postn was found to be significantly upregulated in skeletal muscle and myoblasts of DM1 patients. Next, we knocked down Postn using a short hairpin RNA (shRNA) in DM1 myoblast cells and found that the myogenesis defects in the DM1 group were successfully rescued, as evidenced by increases in the myotube area, the fusion index, and the expression of myogenesis regulatory genes. Similarly, Postn knockdown in normal myoblast cells enhanced myogenesis. As POSTN is a secreted protein, we treated the DM1 myoblast cells with a POSTN-neutralizing antibody and found that DM1 myogenesis defects were successfully rescued by POSTN neutralization. We also tested the myogenic ability of myoblasts in the skeletal muscle injury mouse model and found that Postn knockdown improved the myogenic ability of DM1 myoblasts. The activity of the TGF-β/Smad3 pathway was upregulated during DM1 myogenesis but repressed when inhibiting Postn with a Postn shRNA or a POSTN-neutralizing antibody, which suggested that the TGF-β/Smad3 pathway might mediate the function of Postn in DM1 myogenesis. These results suggest that Postn is a potential therapeutical target for the treatment of myogenesis defects in DM1.
Collapse
Affiliation(s)
- Xiaopeng Shen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Zhongxian Liu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Chunguang Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Feng Xu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Jingyi Zhang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Meng Li
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yang Lei
- Wuhu Center for Disease Control and Prevention, Wuhu, China
| | - Ao Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Chao Bi
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
17
|
Kölbel H, Preuße C, Brand L, von Moers A, Della Marina A, Schuelke M, Roos A, Goebel HH, Schara-Schmidt U, Stenzel W. Inflammation, fibrosis and skeletal muscle regeneration in LGMDR9 are orchestrated by macrophages. Neuropathol Appl Neurobiol 2021; 47:856-866. [PMID: 33973272 DOI: 10.1111/nan.12730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/03/2021] [Accepted: 05/01/2021] [Indexed: 11/29/2022]
Abstract
AIMS Variable degrees of inflammation, necrosis, regeneration and fibrofatty replacement are part of the pathological spectrum of the dystrophic process in alpha dystroglycanopathy LGMDR9 (FKRP-related, OMIM #607155), one of the most prevailing types of LGMDs worldwide. Inflammatory processes and their complex interplay with vascular, myogenic and mesenchymal cells may have a major impact on disease development. The purpose of our study is to describe the specific immune morphological features in muscle tissue of patients with LGMDR9 to enable a better understanding of the phenotype of muscle damage leading to disease progression. METHODS We have analysed skeletal muscle biopsies of 17 patients genetically confirmed as having LGMDR9 by histopathological and molecular techniques. RESULTS We identified CD206+ MHC class II+ and STAT6+ immune-repressed macrophages dominating the endomysial infiltrate in areas of myofibre regeneration and fibrosis. Additionally, PDGFRβ+ pericytes were located around MHC class II+ activated capillaries residing in close proximity to areas of fibrosis and regenerating fibres. Expression of VEGF was found on many regenerating neonatal myosin+ fibres, myofibres and CD206+ macrophages also co-expressed VEGF. CONCLUSION Our results show characteristic immune inflammatory features in LGMDR9 and more specifically shed light on the predominant role of macrophages and their function in vascular organisation, fibrosis and myogenesis. Understanding disease-specific immune phenomena potentially inform about possibilities for anti-fibrotic and anti-inflammatory therapeutic strategies, which may complement Ribitol replacement and gene therapies for LGMDR9 that may be available in the future.
Collapse
Affiliation(s)
- Heike Kölbel
- Department of Neuropaediatrics, Neuromuscular Centre, Universitätsmedizin Essen, Germany
| | - Corinna Preuße
- Department of Neuropathology, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Münster, Germany
| | - Lukas Brand
- Department of Neuropaediatrics, Neuromuscular Centre, Universitätsmedizin Essen, Germany
| | - Arpad von Moers
- Department of Paediatrics and Neuropaediatrics, DRK Klinikum Westend, Berlin, Germany
| | - Adela Della Marina
- Department of Neuropaediatrics, Neuromuscular Centre, Universitätsmedizin Essen, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Roos
- Department of Neuropaediatrics, Neuromuscular Centre, Universitätsmedizin Essen, Germany
| | - Hans-Hilmar Goebel
- Department of Neuropathology, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neuropathology, Universitätsmedizin Mainz, Germany
| | - Ulrike Schara-Schmidt
- Department of Neuropaediatrics, Neuromuscular Centre, Universitätsmedizin Essen, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
18
|
Periostin Is Required for the Maintenance of Muscle Fibers during Muscle Regeneration. Int J Mol Sci 2021; 22:ijms22073627. [PMID: 33807264 PMCID: PMC8036386 DOI: 10.3390/ijms22073627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 02/03/2023] Open
Abstract
Skeletal muscle regeneration is a well-organized process that requires remodeling of the extracellular matrix (ECM). In this study, we revealed the protective role of periostin, a matricellular protein that binds to several ECM proteins during muscle regeneration. In intact muscle, periostin was localized at the neuromuscular junction, muscle spindle, and myotendinous junction, which are connection sites between muscle fibers and nerves or tendons. During muscle regeneration, periostin exhibited robustly increased expression and localization at the interstitial space. Periostin-null mice showed decreased muscle weight due to the loss of muscle fibers during repeated muscle regeneration. Cultured muscle progenitor cells from periostin-null mice showed no deficiencies in their proliferation, differentiation, and the expression of Pax7, MyoD, and myogenin, suggesting that the loss of muscle fibers in periostin-null mice was not due to the impaired function of muscle stem/progenitor cells. Periostin-null mice displayed a decreased number of CD31-positive blood vessels during muscle regeneration, suggesting that the decreased nutritional supply from blood vessels was the cause of muscle fiber loss in periostin-null mice. These results highlight the novel role of periostin in maintaining muscle mass during muscle regeneration.
Collapse
|
19
|
Jacobson KR, Lipp S, Acuna A, Leng Y, Bu Y, Calve S. Comparative Analysis of the Extracellular Matrix Proteome across the Myotendinous Junction. J Proteome Res 2020; 19:3955-3967. [PMID: 32830507 DOI: 10.1021/acs.jproteome.0c00248] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The myotendinous junction is a highly interdigitated interface designed to transfer muscle-generated force to tendon. Understanding how this interface is formed and organized, as well as identifying tendon- and muscle-specific extracellular matrix (ECM), is critical for designing effective regenerative therapies to restore functionality to damaged muscle-tendon units. However, a comparative analysis of the ECM proteome across this interface has not been conducted. The goal of this study was to resolve the distribution of ECM proteins that are uniformly expressed as well as those specific to each of the muscle, tendon, and junction tissues. The soleus muscles from 5-month-old wild-type C57BL/6 mice were harvested and dissected into the central muscle (M) away from tendon, the junction between muscle and tendon (J) and the tendon (T). Tissues were processed by either homogenizing in guanidine hydrochloride or fractionating to isolate the ECM from more soluble intracellular components and then analyzed using liquid chromatography-tandem mass spectrometry. Overall, we found that both tissue processing methods generated similar ECM profiles. Many ECM were found across the muscle-tendon unit, including type I collagen and associated fibril-regulating proteins. The ECM identified exclusively in M were primarily related to the basal lamina, whereas those specific to T and J tissue included thrombospondins and other matricellular ECM. Type XXII collagen (COL22A1) was restricted to J, and we identified COL5A3 as a potential marker of the muscle-tendon interface. Immunohistochemical analysis of key proteins confirmed the restriction of some basal lamina proteins to M, tenascin-C to T, and COL22A1 to J. COL5A3, PRELP, and POSTN were visualized in the tissue surrounding the junction, suggesting that these proteins play a role in stabilizing the interface. This comparative map provides a guide for tissue-specific ECM that can facilitate the spatial visualization of M, J, and T tissues and inform musculoskeletal regenerative therapies.
Collapse
Affiliation(s)
- Kathryn R Jacobson
- Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, Indiana 47907, United States
| | - Sarah Lipp
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - Andrea Acuna
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - Yue Leng
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - Ye Bu
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - Sarah Calve
- Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, Indiana 47907, United States.,Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States.,Paul M. Rady Department of Mechanical Engineering, University of Colorado-Boulder, 1111 Engineering Center, 427 UCB, Boulder, Colorado 80309, United States
| |
Collapse
|
20
|
Ceafalan LC, Dobre M, Milanesi E, Niculae AM, Manole E, Gherghiceanu M, Hinescu ME. Gene expression profile of adhesion and extracellular matrix molecules during early stages of skeletal muscle regeneration. J Cell Mol Med 2020; 24:10140-10150. [PMID: 32681815 PMCID: PMC7520258 DOI: 10.1111/jcmm.15624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle regeneration implies the coordination of myogenesis with the recruitment of myeloid cells and extracellular matrix (ECM) remodelling. Currently, there are no specific biomarkers to diagnose the severity and prognosis of muscle lesions. In order to investigate the gene expression profile of extracellular matrix and adhesion molecules, as premises of homo‐ or heterocellular cooperation and milestones for skeletal muscle regeneration, we performed a gene expression analysis for genes involved in cellular cooperation, migration and ECM remodelling in a mouse model of acute crush injury. The results obtained at two early time‐points post‐injury were compared to a GSE5413 data set from two other trauma models. Third day post‐injury, when inflammatory cells invaded, genes associated with cell‐matrix interactions and migration were up‐regulated. After day 5, as myoblast migration and differentiation started, genes for basement membrane constituents were found down‐regulated, whereas genes for ECM molecules, macrophage, myoblast adhesion, and migration receptors were up‐regulated. However, the profile and the induction time varied according to the experimental model, with only few genes being constantly up‐regulated. Gene up‐regulation was higher, delayed and more diverse following more severe trauma. Moreover, one of the most up‐regulated genes was periostin, suggestive for severe muscle damage and unfavourable architecture restoration.
Collapse
Affiliation(s)
- Laura C Ceafalan
- Cell Biology, Neurosciences and Experimental Myology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania.,Department of Cellular and Molecular Biology and Histology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - Maria Dobre
- Molecular Pathology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania
| | - Elena Milanesi
- Molecular Pathology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania.,Radiobiology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania
| | - Andrei M Niculae
- Department of Cellular and Molecular Biology and Histology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - Emilia Manole
- Cell Biology, Neurosciences and Experimental Myology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania
| | - Mihaela Gherghiceanu
- Department of Cellular and Molecular Biology and Histology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.,Ultrastructural Pathology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania
| | - Mihail E Hinescu
- Cell Biology, Neurosciences and Experimental Myology Laboratory, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania.,Department of Cellular and Molecular Biology and Histology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
21
|
Kerschan-Schindl K, Tiefenböck TM, Föger-Samwald U, Payr S, Frenzel S, Marculescu R, Gleiss A, Sarahrudi K, Pietschmann P. Circulating Myostatin Levels Decrease Transiently after Implantation of a Hip Hemi-Arthroplasty. Gerontology 2020; 66:393-400. [PMID: 32454508 DOI: 10.1159/000507731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/03/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Muscle and bone metabolism are both important for the healing of fractures and the regeneration of injured muscle tissue. The aim of this investigation was to evaluate myostatin and other regulating factors in patients with hip fractures who underwent hemi-arthroplasty. METHODS Serum levels of myostatin (MSTN), follistatin (FSTN), dickkopf-1 (Dkk1), and periostin (PSTN) as well as markers of bone turnover were evaluated in patients with hip fractures before surgery and twice in the 2 weeks after surgery. These parameters were also evaluated in age- and gender-matched subjects without major musculoskeletal injury. RESULTS MSTN was transiently reduced; its opponent FSTN was transiently increased. Dkk1, the negative regulator of bone mass, and PSTN, a marker of subperiosteal bone formation, increased after surgery. With regard to markers of bone turnover, resorption was elevated during the entire period of observation whereas the early bone formation marker N-terminal propeptide of type I collagen was elevated 12 days after surgery. CONCLUSIONS Unexpectedly, MSTN, a negative regulator of muscle growth, was reduced after surgery compared with before surgery. As musculoskeletal markers are altered during bone healing, they do not reflect general bone metabolism after fracture or joint arthroplasty. This is important because many elderly patients receive treatment for osteoporosis.
Collapse
Affiliation(s)
- Katharina Kerschan-Schindl
- Department of Physical Medicine, Rehabilitation and Occupational Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas M Tiefenböck
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Ursula Föger-Samwald
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Stephan Payr
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Frenzel
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Center of Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Kambiz Sarahrudi
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria, .,Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria,
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|