1
|
Han J, Zhang J, Zhang X, Luo W, Liu L, Zhu Y, Liu Q, Zhang XA. Emerging role and function of Hippo-YAP/TAZ signaling pathway in musculoskeletal disorders. Stem Cell Res Ther 2024; 15:386. [PMID: 39468616 PMCID: PMC11520482 DOI: 10.1186/s13287-024-04011-9] [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: 08/06/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Hippo pathway is an evolutionarily conservative key pathway that regulates organ size and tissue regeneration by regulating cell proliferation, differentiation and apoptosis. Yes-associated protein 1 (YAP)/ WW domain-containing transcription regulator 1 (TAZ) serves as a pivotal transcription factor within the Hippo signaling pathway, which undergoes negative regulation by the Hippo pathway. The expression of YAP/TAZ affects various biological processes, including differentiation of osteoblasts (OB) and osteoclasts (OC), cartilage homeostasis, skeletal muscle development, regeneration and quality maintenance. At the same time, the dysregulation of the Hippo pathway can concurrently contribute to the development of various musculoskeletal disorders, including bone tumors, osteoporosis (OP), osteoarthritis (OA), intervertebral disc degeneration (IDD), muscular dystrophy, and rhabdomyosarcoma (RMS). Therefore, targeting the Hippo pathway has emerged as a promising therapeutic strategy for the treatment of musculoskeletal disorders. The focus of this review is to elucidate the mechanisms by which the Hippo pathway maintains homeostasis in bone, cartilage, and skeletal muscle, while also providing a comprehensive summary of the pivotal role played by core components of this pathway in musculoskeletal diseases. The efficacy and feasibility of Hippo pathway-related drugs for targeted therapy of musculoskeletal diseases are also discussed in our study. These endeavors offer novel insights into the application of Hippo signaling in musculoskeletal disorders, providing effective therapeutic targets and potential drug candidates for treating such conditions.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xiaoyi Zhang
- College of Second Clinical Medical, China Medical University, Shenyang, 110122, China
| | - Wenxin Luo
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Lifei Liu
- Department of Rehabilitation, The People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Yuqing Zhu
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Qingfeng Liu
- Department of General Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China.
| |
Collapse
|
2
|
Delaney R, O'Halloran KD. Respiratory performance in Duchenne muscular dystrophy: Clinical manifestations and lessons from animal models. Exp Physiol 2024; 109:1426-1445. [PMID: 39023735 PMCID: PMC11363095 DOI: 10.1113/ep091967] [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: 04/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal genetic neuromuscular disease. Lack of dystrophin in skeletal muscles leads to intrinsic weakness, injury, subsequent degeneration and fibrosis, decreasing contractile function. Dystropathology eventually presents in all inspiratory and expiratory muscles of breathing, severely curtailing their critical function. In people with DMD, premature death is caused by respiratory or cardiac failure. There is an urgent need to develop therapies that improve quality of life and extend life expectancy in DMD. Surprisingly, there is a dearth of information on respiratory control in animal models of DMD, and respiratory outcome measures are often limited or absent in clinical trials. Characterization of respiratory performance in murine and canine models has revealed extensive remodelling of the diaphragm, the major muscle of inspiration. However, significant compensation by extradiaphragmatic muscles of breathing is evident in early disease, contributing to preservation of peak respiratory system performance. Loss of compensation afforded by accessory muscles in advanced disease is ultimately associated with compromised respiratory performance. A new and potentially more translatable murine model of DMD, the D2.mdx mouse, has recently been developed. Respiratory performance in D2.mdx mice is yet to be characterized fully. However, based on histopathological features, D2.mdx mice might serve as useful preclinical models, facilitating the testing of new therapeutics that rescue respiratory function. This review summarizes the pathophysiological mechanisms associated with DMD both in humans and in animal models, with a focus on breathing. We consider the translational value of each model to human DMD and highlight the urgent need for comprehensive characterization of breathing in representative preclinical models to better inform human trials.
Collapse
|
3
|
Ren T, Xu M, Lin W, Luo W, Zhang X. Transcriptome sequencing reveals the potential mechanisms of dietary lutein regulation on chicken leg muscle development. Poult Sci 2024; 103:104265. [PMID: 39293263 PMCID: PMC11426042 DOI: 10.1016/j.psj.2024.104265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/03/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024] Open
Abstract
Lutein is an antioxidant that can indicate the oxidative status of organisms through its coloration and may be involved in the development process of chicken skeletal muscle. In this study, after feeding Nanhai Yellow Chickens with lutein-containing feed for 21 d, the lutein group significantly increased the muscle fiber diameter and decreased the fiber density in the chicken's leg muscles compared to the control group. To elucidate the potential regulatory mechanisms by which lutein is involved in muscle development, RNA-seq was used to detect changes in gene expression in chicken leg muscle tissue. After data analysis, a total of 249 significantly differentially expressed genes (DEG) were identified, including TGF-β superfamily (MSTN and TGFB1) and nonreceptor tyrosine kinase c-Src (SRC). Results from GO and KEGG analysis showed that the DEGs were enriched in GO terms such as positive regulation of the ERK1/ERK2 cascade and negative regulation of myoblast differentiation, as well as signaling pathways including the Toll-like receptor signaling pathway and the MAPK signaling pathway. These significantly enriched GO terms and pathways are closely related to muscle development, suggesting that lutein may play an important role in the process of chicken muscle development. This study provides insights into the regulatory mechanisms of dietary lutein on chicken muscle development.
Collapse
Affiliation(s)
- Tuanhui Ren
- Department of Animal Genetics, College of Animal Science, Breeding and Reproduction, South China Agricultural University, Guangzhou, 510642, China; Guangdong Key Laboratory of Genome and Molecular Breeding of Agricultural Animals and Key Laboratory of Chicken Genetic Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Meng Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Wujian Lin
- Department of Animal Genetics, College of Animal Science, Breeding and Reproduction, South China Agricultural University, Guangzhou, 510642, China; Guangdong Key Laboratory of Genome and Molecular Breeding of Agricultural Animals and Key Laboratory of Chicken Genetic Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wen Luo
- Department of Animal Genetics, College of Animal Science, Breeding and Reproduction, South China Agricultural University, Guangzhou, 510642, China; Guangdong Key Laboratory of Genome and Molecular Breeding of Agricultural Animals and Key Laboratory of Chicken Genetic Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Xiquan Zhang
- Department of Animal Genetics, College of Animal Science, Breeding and Reproduction, South China Agricultural University, Guangzhou, 510642, China; Guangdong Key Laboratory of Genome and Molecular Breeding of Agricultural Animals and Key Laboratory of Chicken Genetic Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
4
|
Yan X, Fu P, Zhang Y, Ling D, Reynolds L, Hua W, Wang Z, Ma F, Li B, Yu J, Liu Y, Gong L, Zhang E. MCC950 Ameliorates Diabetic Muscle Atrophy in Mice by Inhibition of Pyroptosis and Its Synergistic Effect with Aerobic Exercise. Molecules 2024; 29:712. [PMID: 38338456 PMCID: PMC10856337 DOI: 10.3390/molecules29030712] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetic muscle atrophy is an inflammation-related complication of type-2 diabetes mellitus (T2DM). Even though regular exercise prevents further deterioration of atrophic status, there is no effective mediator available for treatment and the underlying cellular mechanisms are less explored. In this study, we investigated the therapeutic potential of MCC950, a specific, small-molecule inhibitor of NLRP3, to treat pyroptosis and diabetic muscle atrophy in mice. Furthermore, we used MCC950 to intervene in the protective effects of aerobic exercise against muscle atrophy in diabetic mice. Blood and gastrocnemius muscle (GAS) samples were collected after 12 weeks of intervention and the atrophic state was assessed. We initially corroborated a diabetic muscle atrophy phenotype in db/db mice (D) by comparison with control m/m mice (W) by examining parameters such as fasting blood glucose (D vs. W: 24.47 ± 0.45 mmol L-1 vs. 4.26 ± 0.6 mmol L-1, p < 0.05), grip strength (D vs. W: 166.87 ± 15.19 g vs. 191.76 ± 14.13 g, p < 0.05), exercise time (D vs. W: 1082.38 ± 104.67 s vs. 1716 ± 168.55 s, p < 0.05) and exercise speed to exhaustion (D vs. W: 24.25 ± 2.12 m min-1 vs. 34.75 ± 2.66 m min-1, p < 0.05), GAS wet weight (D vs. W: 0.07 ± 0.01 g vs. 0.13 ± 0.01 g, p < 0.05), the ratio of GAS wet weight to body weight (D vs. W: 0.18 ± 0.01% vs. 0.54 ± 0.02%, p < 0.05), and muscle fiber cross-sectional area (FCSA) (D vs. W: 1875 ± 368.19 µm2 vs. 2747.83 ± 406.44 µm2, p < 0.05). We found that both MCC950 (10 mg kg-1) treatment and exercise improved the atrophic parameters that had deteriorated in the db/db mice, inhibited serum inflammatory markers and significantly attenuated pyroptosis in atrophic GAS. In addition, a combined MCC950 treatment with exercise (DEI) exhibited a further improvement in glucose uptake capacity and muscle performance. This combined treatment also improved the FCSA of GAS muscle indicated by Laminin immunofluorescence compared to the group with the inhibitor treatment alone (DI) (DEI vs. DI: 2597 ± 310.97 vs. 1974.67 ± 326.15 µm2, p < 0.05) or exercise only (DE) (DEI vs. DE: 2597 ± 310.97 vs. 2006.33 ± 263.468 µm2, p < 0.05). Intriguingly, the combination of MCC950 treatment and exercise significantly reduced NLRP3-mediated inflammatory factors such as cleaved-Caspase-1, GSDMD-N and prevented apoptosis and pyroptosis in atrophic GAS. These findings for the first time demonstrate that targeting NLRP3-mediated pyroptosis with MCC950 improves diabetic muscle homeostasis and muscle function. We also report that inhibiting pyroptosis by MCC950 can enhance the beneficial effects of aerobic exercise on diabetic muscle atrophy. Since T2DM and muscle atrophy are age-related diseases, the young mice used in the current study do not seem to fully reflect the characteristics of diabetic muscle atrophy. Considering the fragile nature of db/db mice and for the complete implementation of the exercise intervention, we used relatively young db/db mice and the atrophic state in the mice was thoroughly confirmed. Taken together, the current study comprehensively investigated the therapeutic effect of NLRP3-mediated pyroptosis inhibited by MCC950 on diabetic muscle mass, strength and exercise performance, as well as the synergistic effects of MCC950 and exercise intervention, therefore providing a novel strategy for the treatment of the disease.
Collapse
Affiliation(s)
- Xiaoyu Yan
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Pengyu Fu
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- Department of Physical Education, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yimin Zhang
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Dongmei Ling
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Lewis Reynolds
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, 21428 Malmö, Sweden (E.Z.)
- NanoLund Center for NanoScience, Lund University, 22100 Lund, Sweden
| | - Weicheng Hua
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Zhiyuan Wang
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Fangyuan Ma
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- School of Life Sciences, Nankai University, Tianjin 300071, China
| | - Boxuan Li
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Jingjing Yu
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
| | - Yujia Liu
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- Institute of Physical Education, Jiangsu Normal University, Xuzhou 221116, China
| | - Lijing Gong
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, 21428 Malmö, Sweden (E.Z.)
- NanoLund Center for NanoScience, Lund University, 22100 Lund, Sweden
| |
Collapse
|
5
|
Kim HJ, Kim H, Lee JH, Hwangbo C. Toll-like receptor 4 (TLR4): new insight immune and aging. Immun Ageing 2023; 20:67. [PMID: 38001481 PMCID: PMC10668412 DOI: 10.1186/s12979-023-00383-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023]
Abstract
TLR4, a transmembrane receptor, plays a central role in the innate immune response. TLR4 not only engages with exogenous ligands at the cellular membrane's surface but also interacts with intracellular ligands, initiating intricate intracellular signaling cascades. Through MyD88, an adaptor protein, TLR4 activates transcription factors NF-κB and AP-1, thereby facilitating the upregulation of pro-inflammatory cytokines. Another adapter protein linked to TLR4, known as TRIF, autonomously propagates signaling pathways, resulting in heightened interferon expression. Recently, TLR4 has garnered attention as a significant factor in the regulation of symptoms in aging-related disorders. The persistent inflammatory response triggered by TLR4 contributes to the onset and exacerbation of these disorders. In addition, alterations in TLR4 expression levels play a pivotal role in modifying the manifestations of age-related diseases. In this review, we aim to consolidate the impact of TLR4 on cellular senescence and aging-related ailments, highlighting the potential of TLR4 as a novel therapeutic target that extends beyond immune responses.
Collapse
Affiliation(s)
- Hyo-Jin Kim
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyemin Kim
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry (BK21 Four), College of Natural Sciences, Kangwon National University, Chuncheon, 24414, Republic of Korea
| | - Cheol Hwangbo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
6
|
Li L, Huang C, Pang J, Huang Y, Chen X, Chen G. Advances in research on cell models for skeletal muscle atrophy. Biomed Pharmacother 2023; 167:115517. [PMID: 37738794 DOI: 10.1016/j.biopha.2023.115517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
Skeletal muscle, the largest organ in the human body, plays a crucial role in supporting and defending the body and is essential for movement. It also participates in regulating the processes of protein synthesis and degradation. Inhibition of protein synthesis and activation of degradation metabolism can both lead to the development of skeletal muscle atrophy, a pathological condition characterized by a decrease in muscle mass and fiber size. Many physiological and pathological conditions can cause a decline in muscle mass, but the underlying mechanisms of its pathogenesis remain incompletely understood, and the selection of treatment strategies and efficacy evaluations vary. Moreover, the early symptoms of this condition are often not apparent, making it easily overlooked in clinical practice. Therefore, it is necessary to develop and use cell models to understand the etiology and influencing factors of skeletal muscle atrophy. In this review, we summarize the methods used to construct skeletal muscle cell models, including hormone, inflammation, cachexia, genetic engineering, drug, and physicochemical models. We also analyze, compare, and evaluate the various construction and assessment methods.
Collapse
Affiliation(s)
- Liwei Li
- Guangdong Medical University, Wenming East Road 2, Xiashan District, Zhanjiang 524000, Guangdong, China
| | - Chunman Huang
- Guangdong Medical University, Wenming East Road 2, Xiashan District, Zhanjiang 524000, Guangdong, China
| | - Jingqun Pang
- Guangdong Medical University, Wenming East Road 2, Xiashan District, Zhanjiang 524000, Guangdong, China
| | - Yongbin Huang
- Guangdong Medical University, Wenming East Road 2, Xiashan District, Zhanjiang 524000, Guangdong, China
| | - Xinxin Chen
- Institute of Health Promotion and Medical Communication Studies, Affliated Hospital of Guangdong Medical University, South Renmin Road 57, Xiashan District, Zhanjiang 524000, Guangdong, China
| | - Guanghua Chen
- Orthopaedic Center, Affliated Hospital of Guangdong Medical University, South Renmin Road 57, Xiashan District, Zhanjiang 524000, Guangdong, China.
| |
Collapse
|
7
|
Boonlaos A, Uddin MJ, Temyord K, Jattawa D, Kayan A. Muscle fiber characteristics and expression level of Troponin T3, Toll-like receptor 2, and Toll-like receptor 4 genes in chicken meat with white striping. Vet World 2023; 16:1415-1420. [PMID: 37621550 PMCID: PMC10446722 DOI: 10.14202/vetworld.2023.1415-1420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/31/2023] [Indexed: 08/26/2023] Open
Abstract
Background and Aim The poultry industry faces an emerging muscular defect in chicken meat called white striping (WS). The biological processes associated with WS myopathy are immune system activation, angiogenesis, hypoxia, cell death, and striated muscle contraction. We examined the Troponin T3 (TNNT3), Toll-like receptor 2 (TLR2), and Toll-like receptor 4 (TLR4) genes based on their functions related to muscle contraction and the innate immune system. This study aimed to determine the muscle fiber characteristics (MFCs) and expression level of TNNT3, TLR2, and TLR4 genes in white striping chicken meat (WSCM). Materials and Methods A total of 428 breast samples were randomly collected from a commercial poultry processing plant. The samples were classified into four levels: 0 (normal), 1 (moderate WS), 2 (severe WS), and 3 (extreme WS). Five samples per group were selected to evaluate MFCs, including total number of muscle fibers, muscle fiber diameter, cross-sectional area, endomysium thickness, and perimysium thickness. Five samples per group were selected for ribonucleic acid (RNA) isolation to evaluate the messenger RNA (mRNA) expression levels of TNNT3, TLR2, and TLR4 genes related to WS. Results Statistical analysis revealed that the total number of fibers, endomysium thickness, and perimysium thickness significantly differed between groups (p < 0.05). Muscle fiber diameter and cross-sectional area did not significantly differ (p > 0.05). The expression of the TNNT3 gene did not significantly differ among groups (p > 0.05). Toll-like receptor 2 and TLR4 mRNA expression significantly differed among groups (p < 0.05). Conclusion These detailed MFCs will provide baseline information to observe WS in chicken meat. Toll-like receptor 2 and TLR4 genes may play a role in the occurrence of WS in chicken meat through non-specific immune reactions.
Collapse
Affiliation(s)
- Antika Boonlaos
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| | - Muhammad Jasim Uddin
- School of Veterinary Medicine, Murdoch University, Western Australia, Australia
- Center for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, Australia
| | - Katchaporn Temyord
- Bureau of Livestock Standard and Certification, Department of Livestock Development, Bangkok, Thailand
| | - Danai Jattawa
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| | - Autchara Kayan
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
8
|
Inflammaging: Implications in Sarcopenia. Int J Mol Sci 2022; 23:ijms232315039. [PMID: 36499366 PMCID: PMC9740553 DOI: 10.3390/ijms232315039] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
In a world in which life expectancy is increasing, understanding and promoting healthy aging becomes a contemporary demand. In the elderly, a sterile, chronic and low-grade systemic inflammation known as "inflammaging" is linked with many age-associated diseases. Considering sarcopenia as a loss of strength and mass of skeletal muscle related to aging, correlations between these two terms have been proposed. Better knowledge of the immune system players in skeletal muscle would help to elucidate their implications in sarcopenia. Characterizing the activators of damage sensors and the downstream effectors explains the inference with skeletal muscle performance. Sarcopenia has also been linked to chronic diseases such as diabetes, metabolic syndrome and obesity. Implications of inflammatory signals from these diseases negatively affect skeletal muscle. Autophagic mechanisms are closely related with the inflammasome, as autophagy eliminates stress signaling sent by damage organelles, but also acts with an immunomodulatory function affecting immune cells and cytokine release. The use of melatonin, an antioxidant, ROS scavenger and immune and autophagy modulator, or senotherapeutic compounds targeting senescent cells could represent strategies to counteract inflammation. This review aims to present the many factors regulating skeletal muscle inflammaging and their major implications in order to understand the molecular mechanisms involved in sarcopenia.
Collapse
|
9
|
PD-1 Alleviates Cisplatin-Induced Muscle Atrophy by Regulating Inflammation and Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11091839. [PMID: 36139912 PMCID: PMC9495887 DOI: 10.3390/antiox11091839] [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: 07/25/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 12/03/2022] Open
Abstract
Skeletal muscle atrophy is an important characteristic of cachexia, which can be induced by chemotherapy and significantly contributes to functional muscle impairment. Inflammation and oxidative stress are believed to play important roles in the muscle atrophy observed in cachexia, but whether programmed cell death protein 1 (PD-1) is affected by this condition remains unclear. PD-1 is a membrane protein that is expressed on the surface of many immune cells and plays an important role in adaptive immune responses and autoimmunity. Thus, we investigated the role and underlying mechanism of PD-1 in cisplatin-induced muscle atrophy in mice. We found that PD-1 knockout dramatically contributed to skeletal muscle atrophy. Mechanistically, we found that E3 ubiquitin-protein ligases were significantly increased in PD-1 knockout mice after cisplatin treatment. In addition, we found that PD-1 knockout significantly exacerbated cisplatin-induced skeletal muscle inflammation and oxidative stress. Moreover, we found that there were significant increases in ferroptosis-related and autophagy-related genes in PD-1 knockout mice after cisplatin treatment. These data indicate that PD-1 plays an important role in cisplatin-induced skeletal muscle atrophy.
Collapse
|
10
|
Han X, Han J, Wang N, Ji G, Guo R, Li J, Wu H, Ma S, Fang P, Song X. Identification of Auxiliary Biomarkers and Description of the Immune Microenvironmental Characteristics in Duchenne Muscular Dystrophy by Bioinformatical Analysis and Experiment. Front Neurosci 2022; 16:891670. [PMID: 35720684 PMCID: PMC9204148 DOI: 10.3389/fnins.2022.891670] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a genetic muscle disorder characterized by progressive muscle wasting associated with persistent inflammation. In this study, we aimed to identify auxiliary biomarkers and further characterize the immune microenvironment in DMD. Methods Differentially expressed genes (DEGs) were identified between DMD and normal muscle tissues based on Gene Expression Omnibus (GEO) datasets. Bioinformatical analysis was used to screen and identify potential diagnostic signatures of DMD which were further validated by real-time quantitative reverse transcription PCR (RT-qPCR). We also performed single-sample gene-set enrichment analysis (ssGSEA) to characterize the proportion of tissue-infiltrating immune cells to determine the inflammatory state of DMD. Results In total, 182 downregulated genes and 263 upregulated genes were identified in DMD. C3, SPP1, TMSB10, TYROBP were regarded as adjunct biomarkers and successfully validated by RT-qPCR. The infiltration of macrophages, CD4+, and CD8+ T cells was significantly higher (p < 0.05) in DMD compared with normal muscle tissues, while the infiltration of activated B cells, CD56dim natural killer cells, and type 17 T helper (Th17) cells was lower. In addition, the four biomarkers (C3, SPP1, TMSB10, TYROBP) were strongly associated with immune cells and immune-related pathways in DMD muscle tissues. Conclusion Analyses demonstrated C3, SPP1, TMSB10, and TYROBP may serve as biomarkers and enhance our understanding of immune responses in DMD. The infiltration of immune cells into the muscle microenvironment might exert a critical impact on the development and occurrence of DMD.
Collapse
Affiliation(s)
- Xu Han
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Jingzhe Han
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Ning Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Guang Ji
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Ruoyi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Jing Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Hongran Wu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Shaojuan Ma
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Pingping Fang
- Department of Neurology, Handan Central Hospital, Handan, China
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
- *Correspondence: Xueqin Song,
| |
Collapse
|
11
|
Identification of hub biomarkers and immune cell infiltration in polymyositis and dermatomyositis. Aging (Albany NY) 2022; 14:4530-4555. [PMID: 35609018 PMCID: PMC9186768 DOI: 10.18632/aging.204098] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/12/2022] [Indexed: 12/03/2022]
Abstract
Objective: Polymyositis (PM) and dermatomyositis (DM) are heterogeneous disorders. However, the etiology of PM/DM development has not been thoroughly clarified. Methods: Gene expression data of PM/DM were obtained from Gene Expression Omnibus. We used robust rank aggregation (RRA) to identify differentially expressed genes (DEGs). Gene Ontology functional enrichment and pathway analyses were used to investigate potential functions of the DEGs. Weighted gene co-expression network analysis (WGCNA) was used to establish a gene co-expression network. CIBERSORT was utilized to analyze the pattern of immune cell infiltration in PM/DM. Protein–protein interaction (PPI) network, Venn, and association analyses between core genes and muscle injury were performed to identify hub genes. Receiver operating characteristic analyses were executed to investigate the value of hub genes in the diagnosis of PM/DM, and the results were verified using the microarray dataset GSE48280. Results: Five datasets were included. The RRA integrated analysis identified 82 significant DEGs. Functional enrichment analysis revealed that immune function and the interferon signaling pathway were enriched in PM/DM. WGCNA outcomes identified MEblue and MEturquoise as key target modules in PM/DM. Immune cell infiltration analysis revealed greater macrophage infiltration and lower regulatory T-cell infiltration in PM/DM patients than in healthy controls. PPI network, Venn, and association analyses of muscle injury identified five putative hub genes: TRIM22, IFI6, IFITM1, IFI35, and IRF9. Conclusions: Our bioinformatics analysis identified new genetic biomarkers of the pathogenesis of PM/DM. We demonstrated that immune cell infiltration plays a pivotal part in the occurrence of PM/DM.
Collapse
|
12
|
Diez-Fuertes F, López-Huertas MR, García-Pérez J, Calonge E, Bermejo M, Mateos E, Martí P, Muelas N, Vílchez JJ, Coiras M, Alcamí J, Rodríguez-Mora S. Transcriptomic Evidence of the Immune Response Activation in Individuals With Limb Girdle Muscular Dystrophy Dominant 2 (LGMDD2) Contributes to Resistance to HIV-1 Infection. Front Cell Dev Biol 2022; 10:839813. [PMID: 35646913 PMCID: PMC9136291 DOI: 10.3389/fcell.2022.839813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
LGMDD2 is a rare form of muscular dystrophy characterized by one of the three heterozygous deletions described within the TNPO3 gene that result in the addition of a 15-amino acid tail in the C-terminus.TNPO3 is involved in the nuclear import of splicing factors and acts as a host cofactor for HIV-1 infection by mechanisms not yet deciphered. Further characterization of the crosstalk between HIV-1 infection and LGMDD2 disease may contribute to a better understanding of both the cellular alterations occurring in LGMDD2 patients and the role of TNPO3 in the HIV-1 cycle. To this regard, transcriptome profiling of PBMCs from LGMDD2 patients carrying the deletion c.2771delA in the TNPO3 gene was compared to healthy controls. A total of 545 differentially expressed genes were detected between LGMDD2 patients and healthy controls, with a high representation of G protein-coupled receptor binding chemokines and metallopeptidases among the most upregulated genes in LGMDD2 patients. Plasma levels of IFN-β and IFN-γ were 4.7- and 2.7-fold higher in LGMDD2 patients, respectively. An increase of 2.3-fold in the expression of the interferon-stimulated gene MxA was observed in activated PBMCs from LGMDD2 patients after ex vivo HIV-1 pseudovirus infection. Thus, the analysis suggests a pro-inflammatory state in LGMDD2 patients also described for other muscular dystrophies, that is characterized by the alteration of IL-17 signaling pathway and the consequent increase of metallopeptidases activity and TNF response. In summary, the increase in interferons and inflammatory mediators suggests an antiviral environment and resistance to HIV-1 infection but that could also impair muscular function in LGMDD2 patients, worsening disease evolution. Biomarkers of disease progression and therapeutic strategies based on these genes and mechanisms should be further investigated for this type of muscular dystrophy.
Collapse
Affiliation(s)
- Francisco Diez-Fuertes
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - María Rosa López-Huertas
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Javier García-Pérez
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Esther Calonge
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Mercedes Bermejo
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Elena Mateos
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Pilar Martí
- Neuromuscular Diseases Unit, Neurology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Nuria Muelas
- Neuromuscular Diseases Unit, Neurology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Juan Jesús Vílchez
- Neuromuscular Diseases Unit, Neurology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Mayte Coiras
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - José Alcamí
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Infectious Diseases Unit, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- *Correspondence: José Alcamí, ; Sara Rodríguez-Mora,
| | - Sara Rodríguez-Mora
- AIDS Immunopathogenesis Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- *Correspondence: José Alcamí, ; Sara Rodríguez-Mora,
| |
Collapse
|
13
|
Ducharme JB, McKenna ZJ, Deyhle MR. Exercise mitigates the Toll of muscle atrophy: A narrative review of the effects of exercise on Toll-like receptor-4 in leukocytes and skeletal muscle. Am J Physiol Cell Physiol 2022; 322:C581-C589. [PMID: 35171696 DOI: 10.1152/ajpcell.00005.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Conditions characterized by muscle wasting such as cachexia and sarcopenia are devastating at the individual level, and they place a profound burden on public health. Evidence suggests that inflammation is likely a mechanistic contributor to the pathogenesis of these conditions. One specific molecule, lipopolysaccharide, has gained attention due to its role in initiating inflammation. Toll-like receptor-4 is the primary receptor for lipopolysaccharide and has been shown to be implicit in the downstream proinflammatory response associated with lipopolysaccharide. Importantly, Toll-like receptor-4 is expressed on various cell types throughout the human body such as leukocytes and skeletal muscle fibers and may have site-specific effects that contribute to muscle wasting conditions based on the location in which activation occurs. Accordingly, reducing proinflammatory signaling at these locations may be an effective strategy at mitigating muscle wasting. Regular exercise training is believed to elicit anti-inflammatory adaptations, but the mechanisms by which this occurs are yet to be fully understood. Understanding the mechanisms by which Toll-like receptor-4 activation contributes to muscle wasting and how exercise affects this, may allow for the development of a non-pharmacological therapeutic intervention. Therefore, in this review, we summarize the current understanding of the lipopolysaccharide/Toll-like receptor-4 axis in leukocytes and skeletal muscle fibers on the pathogenesis of muscle wasting conditions and we critically examine the current evidence regarding the effects of exercise on this axis.
Collapse
Affiliation(s)
- Jeremy B Ducharme
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Zachary J McKenna
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Michael R Deyhle
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
14
|
Gu L, Wang Z, Zhang Y, Zhu N, Li J, Yang M, Wang L, Rong S. TLR13 contributes to skeletal muscle atrophy by increasing insulin resistance in chronic kidney disease. Cell Prolif 2022; 55:e13181. [PMID: 35088922 PMCID: PMC8891551 DOI: 10.1111/cpr.13181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 12/25/2022] Open
Abstract
Objectives Insulin resistance in chronic kidney disease (CKD) stimulates muscle wasting, but the molecular processes behind the resistance are undetermined. However, inflammation in skeletal muscle is implicated in the pathogenesis of insulin resistance and cachexia. Toll‐like receptors (TLRs) are known to regulate local innate immune responses, and microarray data have shown that Tlr13 is upregulated in the muscles of mice with CKD, but the relevance is unknown. Materials and Methods We performed in vitro experiments in C2C12 myotubes and constructed a CKD murine model using subtotal nephrectomy to conduct experiments in vivo. Results Tlr13 expression was stimulated in C2C12 myotubes treated with uremic serum. The expression of Tlr13 was also upregulated in the tibialis anterior muscles of mice with CKD. Tlr13 knockdown with siRNAs in skeletal muscle cells decreased insulin resistance despite the inclusion of uremic serum. This led to increased levels of p‐AKT and suppression of protein degradation. Using immunofluorescence staining and coimmunoprecipitation assay, we found that TLR13 recruits IRF3, which activates Irf3 expression, resulting in decreased AKT activity. Moreover, insulin resistance and proteolysis are re‐induced by Irf3 overexpression under Tlr13 deletion. Conclusions Our results indicate that TLR13 is involved in CKD‐mediated insulin resistance in muscle. In catabolic conditions where insulin signaling is impaired, targeting TLR13 may improve insulin sensitivity and prevent muscle atrophy.
Collapse
Affiliation(s)
- Lijie Gu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifang Wang
- Department of Respiration, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yueyue Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayong Li
- Clinical Laboratory Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man Yang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Tripodi L, Molinaro D, Farini A, Cadiao G, Villa C, Torrente Y. Flavonoids and Omega3 Prevent Muscle and Cardiac Damage in Duchenne Muscular Dystrophy Animal Model. Cells 2021; 10:2917. [PMID: 34831140 PMCID: PMC8616158 DOI: 10.3390/cells10112917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/14/2023] Open
Abstract
Nutraceutical products possess various anti-inflammatory, antiarrhythmic, cardiotonic, and antioxidant pharmacological activities that could be useful in preventing oxidative damage, mainly induced by reactive oxygen species. Previously published data showed that a mixture of polyphenols and polyunsaturated fatty acids, mediate an antioxidative response in mdx mice, Duchenne muscular dystrophy animal model. Dystrophic muscles are characterized by low regenerative capacity, fibrosis, fiber necrosis, inflammatory process, altered autophagic flux and inadequate anti-oxidant response. FLAVOmega β is a mixture of flavonoids and docosahexaenoic acid. In this study, we evaluated the role of these supplements in the amelioration of the pathological phenotype in dystrophic mice through in vitro and in vivo assays. FLAVOmega β reduced inflammation and fibrosis, dampened reactive oxygen species production, and induced an oxidative metabolic switch of myofibers, with consequent increase of mitochondrial activity, vascularization, and fatigue resistance. Therefore, we propose FLAVOmega β as food supplement suitable for preventing muscle weakness, delaying inflammatory milieu, and sustaining physical health in patients affected from DMD.
Collapse
Affiliation(s)
| | | | | | | | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Centro Dino Ferrari, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.T.); (D.M.); (A.F.); (G.C.)
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Centro Dino Ferrari, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.T.); (D.M.); (A.F.); (G.C.)
| |
Collapse
|
16
|
Wharton's Jelly-Derived Mesenchymal Stem Cells Reduce Fibrosis in a Mouse Model of Duchenne Muscular Dystrophy by Upregulating microRNA 499. Biomedicines 2021; 9:biomedicines9091089. [PMID: 34572277 PMCID: PMC8469349 DOI: 10.3390/biomedicines9091089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/23/2021] [Indexed: 01/10/2023] Open
Abstract
The aim of this study was to evaluate the therapeutic effects and mechanisms of Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) in an animal model of Duchenne muscular dystrophy (DMD). Mdx mice (3-5 months old) were administered five different doses of WJ-MSCs through their tail veins. A week after injection, grip strength measurements, creatine kinase (CK) assays, immunohistochemistry, and western blots were performed for comparison between healthy mice, mdx control mice, and WJ-MSC-injected mdx mice. WJ-MSCs exerted dose-dependent multisystem therapeutic effects in mdx mice, by decreasing CK, recovering normal behavior, regenerating muscle, and reducing apoptosis and fibrosis in skeletal muscle. We also confirmed that miR-499-5p is significantly downregulated in mdx mice, and that intravenous injection of WJ-MSCs enhanced its expression, leading to anti-fibrotic effects via targeting TGFβR 1 and 3. Thus, WJ-MSCs may represent novel allogeneic "off-the-shelf" cellular products for the treatment of DMD and possibly other muscle disorders.
Collapse
|
17
|
Lai X, Chen J. C-X-C motif chemokine ligand 12: a potential therapeutic target in Duchenne muscular dystrophy. Bioengineered 2021; 12:5428-5439. [PMID: 34424816 PMCID: PMC8806931 DOI: 10.1080/21655979.2021.1967029] [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] [Indexed: 10/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by a mutant dystrophin protein. DMD patients undergo gradual progressive paralysis until death. Chronic glucocorticoid therapy remains one of the main treatments for DMD, despite the significant side effects. However, its mechanisms of action remain largely unknown. We used bioinformatics tools to identify pathogenic genes involved in DMD and glucocorticoid target genes. Two gene expression profiles containing data from DMD patients and healthy controls (GSE38417 and GSE109178) were downloaded for further analysis. Differentially expressed genes (DEGs) between DMD patients and controls were identified using GEO2R, and glucocorticoid target genes were predicted from the Pharmacogenetics and Pharmacogenomics Knowledge Base. Surprisingly, only one gene, CXCL12 (C-X-C motif chemokine ligand 12), was both a glucocorticoid target and a DEG. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, Gene Ontology term enrichment analysis, and gene set enrichment analysis were performed. A protein-protein interaction network was constructed and hub genes identified using the Search Tool for the Retrieval of Interacting Genes (STRING) database and Cytoscape. Enriched pathways involving the DEGs, including CXCL12, were associated with the immune response and inflammation. Levels of CXCL12 and its receptor CXCR4 (C-X-C motif chemokine receptor 4) were increased in X-linked muscular dystrophy (mdx) mice (DMD models) but became significantly reduced after prednisone treatment. Metformin also reduced the expression of CXCL12 and CXCR4 in mdx mice. In conclusion, the CXCL12-CXCR4 pathway may be a potential target for DMD therapy.
Collapse
Affiliation(s)
- Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Chen
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Bortolon JR, Murata GM, Borges L, Weimann E, Silva MBB, Dermargos A, Hatanaka E. Recovery of Diabetic Rats After Physical Exhaustion: Kinetic Alterations in Muscle Inflammation and Muscle-Signaling Proteins to Atrophy and Hypertrophy. Front Physiol 2020; 11:573416. [PMID: 33281615 PMCID: PMC7688783 DOI: 10.3389/fphys.2020.573416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/09/2020] [Indexed: 01/16/2023] Open
Abstract
The complexity of the adaptive response of diabetics to intense exercise is still poorly understood. To optimize exercise interventions in diabetics, the chronology of inflammatory mediators in muscle and the signaling involved in muscle hypertrophy/atrophy must be understood. Herein, we studied the kinetic inflammatory profile and cellular signaling pathways modulated by physical exhaustion after the induction of type 1 diabetes by streptozotocin in rats. Soleus muscle samples were obtained from diabetic and control groups at the following moments: baseline (no exercise); immediately after exhaustive exercise; and at 2 h, 24 h, 48 h, and 72 h after a treadmill exhaustive exercise. Kinetic production of cytokines and kinetic activation of proteins related to muscle synthesis (p70S6K and Akt) and degradation (GSK3, MuRF1, and MAFbx) were measured in the soleus muscle. We observed that the muscle TNF-α (0.9-fold; p = 0.0007), IL-1β (0.8-fold; p = 0.01), IL-6 (0.8-fold; p = 0.0013), L-selectin (1.0-fold; p = 0.0019), and CINC-2α/β (0.9-fold; p = 0.04) levels were higher in almost all stages of the study in the diabetic animals compared with the control group. Our data showed that exhaustive exercise decreased MAFbx expression in diabetic animals compared to the control group in a time-dependent manner. The decreased activation ratios of MAFbx were followed by a decrease in TNF-α, IL-1β, and IL-6 levels. p70S6k phosphorylation was also decreased in the diabetic group compared to the control group after physical exhaustion. Regarding the activation of proteins related to muscle synthesis and degradation, we found that the alterations induced by exhaustive exercise in the diabetic rats might involve pathways related to synthesis and muscle breakdown. Moreover, after an exhaustive exercise session, the recovery of the inflammatory response in the diabetic animals was slower than that in the control rats while the return of inflammatory cytokines to baseline levels was more effective in the diabetic animals.
Collapse
Affiliation(s)
- José Ricardo Bortolon
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| | - Gilson Masahiro Murata
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| | - Leandro Borges
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| | - Eleine Weimann
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| | - Maysa Braga Barros Silva
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| | | | - Elaine Hatanaka
- Instituto de Ciências da Atividade Física e Esporte (ICAFE), Universidade Cruzeiro do Sul, São Paulo, Brazil
| |
Collapse
|
19
|
Bonomo AC, Pinto-Mariz F, Riederer I, Benjamim CF, Butler-Browne G, Mouly V, Savino W. Crosstalk Between Innate and T Cell Adaptive Immunity With(in) the Muscle. Front Physiol 2020; 11:573347. [PMID: 33071827 PMCID: PMC7531250 DOI: 10.3389/fphys.2020.573347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Growing evidence demonstrates a continuous interaction between the immune system and the skeletal muscle in inflammatory diseases of different pathogenetic origins, in dystrophic conditions such as Duchenne Muscular Dystrophy as well as during normal muscle regeneration. Although one component of the innate immunity, the macrophage, has been extensively studied both in disease conditions and during cell or gene therapy strategies aiming at restoring muscular functions, much less is known about dendritic cells and their primary immunological targets, the T lymphocytes. This review will focus on the dendritic cells and T lymphocytes (including effector and regulatory T-cells), emphasizing the potential cross talk between these cell types and their influence on the structure and function of skeletal muscle.
Collapse
Affiliation(s)
- Adriana C Bonomo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fernanda Pinto-Mariz
- Marzagão Gesteira Institute of Pediatrics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ingo Riederer
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, United Kingdom
| | - Claudia F Benjamim
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Program of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, U974, Center for Research in Myology, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, U974, Center for Research in Myology, Paris, France
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|