1
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2024:e13763. [PMID: 39435630 DOI: 10.1111/cpr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- Department of Orthopedics, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- Department of Orthopedics, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
2
|
Epstein SA, Doles JD, Dasgupta A. KLF10: a point of convergence in cancer cachexia. Curr Opin Support Palliat Care 2024; 18:120-125. [PMID: 39007915 PMCID: PMC11293965 DOI: 10.1097/spc.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PURPOSE OF THE REVIEW Cancer-associated cachexia is a wasting syndrome entailing loss in body mass and a shortened life expectancy. There is currently no effective treatment to abrogate this syndrome, which leads to 20-30% of deaths in patients with cancer. While there have been advancements in defining signaling factors/pathways in cancer-induced muscle wasting, targeting the same in the clinic has not been as successful. Krüppel-like factor 10 (KLF10), a transcription factor implicated in muscle regulation, is regulated by the transforming growth factor-beta signaling pathway. This review proposes KLF10 as a potential convergence point of diverse signaling pathways involved in muscle wasting. RECENT FINDINGS KLF10 was discovered as a target of transforming growth factor-beta decades ago but more recently it has been shown that deletion of KLF10 rescues cancer-induced muscle wasting. Moreover, KLF10 has also been shown to bind key atrophy genes associated with muscle atrophy in vitro . SUMMARY There is an elevated need to explore targets in cachexia, which will successfully translate into the clinic. Investigating a convergence point downstream of multiple signaling pathways might hold promise in developing effective therapies for cachexia.
Collapse
Affiliation(s)
- Savannah A Epstein
- Department of Anatomy, Cell Biology and Physiology, Indiana School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
3
|
Tatarenko Y, Li M, Pouletaut P, Kammoun M, Hawse JR, Joumaa V, Herzog W, Chatelin S, Bensamoun SF. Multiscale analysis of Klf10's impact on the passive mechanical properties of murine skeletal muscle. J Mech Behav Biomed Mater 2024; 150:106298. [PMID: 38096609 DOI: 10.1016/j.jmbbm.2023.106298] [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: 01/13/2023] [Revised: 09/19/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024]
Abstract
Skeletal muscle is a hierarchical structure composed of multiple organizational scales. A major challenge in the biomechanical evaluation of muscle relates to the difficulty in evaluating the experimental mechanical properties at the different organizational levels of the same tissue. Indeed, the ability to integrate mechanical properties evaluated at various levels will allow for improved assessment of the entire tissue, leading to a better understanding of how changes at each level evolve over time and/or impact tissue function, especially in the case of muscle diseases. Therefore, the purpose of this study was to analyze a genetically engineered mouse model (Klf10 KO: Krüppel-Like Factor 10 knockout) with known skeletal muscle defects to compare the mechanical properties with wild-type (WT) controls at the three main muscle scales: the macroscopic (whole muscle), microscopic (fiber) and submicron (myofibril) levels. Passive mechanical tests (ramp, relaxation) were performed on two types of skeletal muscle (soleus and extensor digitorum longus (EDL)). Results of the present study revealed muscle-type specific behaviors in both genotypes only at the microscopic scale. Interestingly, loss of Klf10 expression resulted in increased passive properties in the soleus but decreased passive properties in the EDL compared to WT controls. At the submicron scale, no changes were observed between WT and Klf10 KO myofibrils for either muscle; these results demonstrate that the passive property differences observed at the microscopic scale (fiber) are not caused by sarcomere intrinsic alterations but instead must originate outside the sarcomeres, likely in the collagen-based extracellular matrix. The macroscopic scale revealed similar passive mechanical properties between WT and Klf10 KO hindlimb muscles. The present study has allowed for a better understanding of the role of Klf10 on the passive mechanical properties of skeletal muscle and has provided reference data to the literature which could be used by the community for muscle multiscale modeling.
Collapse
Affiliation(s)
- Y Tatarenko
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338, Biomechanics and Bioengineering, Compiègne, France; ICube, CNRS UMR 7357, University of Strasbourg, Strasbourg, France
| | - M Li
- University of Calgary, Faculty of Kinesiology, Human Performance Laboratory, Calgary, Alberta, Canada
| | - P Pouletaut
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338, Biomechanics and Bioengineering, Compiègne, France
| | - M Kammoun
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338, Biomechanics and Bioengineering, Compiègne, France
| | - J R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - V Joumaa
- University of Calgary, Faculty of Kinesiology, Human Performance Laboratory, Calgary, Alberta, Canada
| | - W Herzog
- University of Calgary, Faculty of Kinesiology, Human Performance Laboratory, Calgary, Alberta, Canada
| | - S Chatelin
- ICube, CNRS UMR 7357, University of Strasbourg, Strasbourg, France
| | - S F Bensamoun
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338, Biomechanics and Bioengineering, Compiègne, France.
| |
Collapse
|
4
|
Yaseen U, Hwang S, Park S, Kim SB, Lee HJ, Cha JY. New Insights into the Role of KLF10 in Tissue Fibrosis. Int J Mol Sci 2024; 25:1276. [PMID: 38279278 PMCID: PMC10816924 DOI: 10.3390/ijms25021276] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Fibrosis, characterized by excessive extracellular matrix accumulation, disrupts normal tissue architecture, causes organ dysfunction, and contributes to numerous chronic diseases. This review focuses on Krüppel-like factor 10 (KLF10), a transcription factor significantly induced by transforming growth factor-β (TGF-β), and its role in fibrosis pathogenesis and progression across various tissues. KLF10, initially identified as TGF-β-inducible early gene-1 (TIEG1), is involved in key biological processes including cell proliferation, differentiation, apoptosis, and immune responses. Our analysis investigated KLF10 gene and protein structures, interaction partners, and context-dependent functions in fibrotic diseases. This review highlights recent findings that underscore KLF10 interaction with pivotal signaling pathways, such as TGF-β, and the modulation of gene expression in fibrotic tissues. We examined the dual role of KLF10 in promoting and inhibiting fibrosis depending on tissue type and fibrotic context. This review also discusses the therapeutic potential of targeting KLF10 in fibrotic diseases, based on its regulatory role in key pathogenic mechanisms. By consolidating current research, this review aims to enhance the understanding of the multifaceted role of KLF10 in fibrosis and stimulate further research into its potential as a therapeutic target in combating fibrotic diseases.
Collapse
Affiliation(s)
- Uzma Yaseen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Sangbin Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soo-Bin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Ho-Jae Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Ji-Young Cha
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| |
Collapse
|
5
|
Ma ZG, Yuan YP, Fan D, Zhang X, Teng T, Song P, Kong CY, Hu C, Wei WY, Tang QZ. IRX2 regulates angiotensin II-induced cardiac fibrosis by transcriptionally activating EGR1 in male mice. Nat Commun 2023; 14:4967. [PMID: 37587150 PMCID: PMC10432509 DOI: 10.1038/s41467-023-40639-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
Cardiac fibrosis is a common feature of chronic heart failure. Iroquois homeobox (IRX) family of transcription factors plays important roles in heart development; however, the role of IRX2 in cardiac fibrosis has not been clarified. Here we report that IRX2 expression is significantly upregulated in the fibrotic hearts. Increased IRX2 expression is mainly derived from cardiac fibroblast (CF) during the angiotensin II (Ang II)-induced fibrotic response. Using two CF-specific Irx2-knockout mouse models, we show that deletion of Irx2 in CFs protect against pathological fibrotic remodelling and improve cardiac function in male mice. In contrast, Irx2 gain of function in CFs exaggerate fibrotic remodelling. Mechanistically, we find that IRX2 directly binds to the promoter of the early growth response factor 1 (EGR1) and subsequently initiates the transcription of several fibrosis-related genes. Our study provides evidence that IRX2 regulates the EGR1 pathway upon Ang II stimulation and drives cardiac fibrosis.
Collapse
Affiliation(s)
- Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Peng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Wen-Ying Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.
- Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, 430060, Wuhan, PR China.
| |
Collapse
|
6
|
Hsu YC, Ho C, Shih YH, Ni WC, Li YC, Chang HC, Lin CL. Knockout of KLF10 Ameliorated Diabetic Renal Fibrosis via Downregulation of DKK-1. Molecules 2022; 27:2644. [PMID: 35565995 PMCID: PMC9105565 DOI: 10.3390/molecules27092644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes-induced chronic kidney disease leads to mortality and morbidity and thus poses a great health burden worldwide. Krüppel-like factor 10 (KLF10), a zinc finger-containing transcription factor, regulates numerous cellular functions, such as proliferation, differentiation, and apoptosis. In this study, we explored the effects of KLF10 on diabetes-induced renal disease by using a KLF10 knockout mice model. Knockout of KLF10 obviously diminished diabetes-induced tumor growth factor-β (TGF-β), fibronectin, and type IV collagen expression, as evidenced by immunohistochemical staining. KLF10 knockout also repressed the expression of Dickkopf-1 (DKK-1) and phosphorylated β-catenin in diabetic mice, as evidenced by immunohistochemical staining and Western blot analysis. Quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) revealed that significantly decreased type IV collagen, fibronectin, and DKK-1 existed in KLF10 knockout diabetic mice compared with control diabetic mice. Moreover, knockout of KLF10 reduced the renal fibrosis, as shown by Masson's Trichrome analysis. Overall, the results indicate that depletion of KLF10 ameliorated diabetic renal fibrosis via the downregulation of DKK-1 expression and inhibited TGF-β1 and phosphorylated β-catenin expression. Our findings suggest that KLF10 may be a promising therapeutic choice for the treatment of diabetes-induced renal fibrosis.
Collapse
Affiliation(s)
- Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Cheng Ho
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Ya-Hsueh Shih
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Wen-Chiu Ni
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Yi-Chen Li
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Hsiu-Ching Chang
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (Y.-H.S.); (W.-C.N.); (Y.-C.L.); (H.-C.C.)
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
7
|
Olivera-Bravo S, Bolatto C, Otero Damianovich G, Stancov M, Cerri S, Rodríguez P, Boragno D, Hernández Mir K, Cuitiño MN, Larrambembere F, Isasi E, Alem D, Canclini L, Marco M, Davyt D, Díaz-Amarilla P. Neuroprotective effects of violacein in a model of inherited amyotrophic lateral sclerosis. Sci Rep 2022; 12:4439. [PMID: 35292673 PMCID: PMC8924276 DOI: 10.1038/s41598-022-06470-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive death of motor neurons and muscle atrophy, with defective neuron-glia interplay and emergence of aberrant glial phenotypes having a role in disease pathology. Here, we have studied if the pigment violacein with several reported protective/antiproliferative properties may control highly neurotoxic astrocytes (AbAs) obtained from spinal cord cultures of symptomatic hSOD1G93A rats, and if it could be neuroprotective in this ALS experimental model. At concentrations lower than those reported as protective, violacein selectively killed aberrant astrocytes. Treatment of hSOD1G93A rats with doses equivalent to the concentrations that killed AbAs caused a marginally significant delay in survival, partially preserved the body weight and soleus muscle mass and improved the integrity of the neuromuscular junction. Reduced motor neuron death and glial reactivity was also found and likely related to decreased inflammation and matrix metalloproteinase-2 and -9. Thus, in spite that new experimental designs aimed at extending the lifespan of hSOD1G93A rats are needed, improvements observed upon violacein treatment suggest a significant therapeutic potential that deserves further studies.
Collapse
Affiliation(s)
- Silvia Olivera-Bravo
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay.
| | - Carmen Bolatto
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Histology and Embryology Department, Faculty of Medicine, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - Gabriel Otero Damianovich
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Matías Stancov
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Sofía Cerri
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Histology and Embryology Department, Faculty of Medicine, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - Paola Rodríguez
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Daniela Boragno
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Karina Hernández Mir
- Histology and Embryology Department, Faculty of Medicine, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - María Noel Cuitiño
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Fernanda Larrambembere
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Eugenia Isasi
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Histology and Embryology Department, Faculty of Medicine, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - Diego Alem
- Genetic Department, IIBCE, Montevideo, Uruguay
| | | | - Marta Marco
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Tumoral Biol Area, Clin Biochem Department, Faculty of Chemistry, UdelaR, Montevideo, Uruguay
| | - Danilo Davyt
- Pharm Chem Lab, Organic Chemistry Department, Faculty of Chemistry, UdelaR, Montevideo, Uruguay
| | - Pablo Díaz-Amarilla
- Cell and Mol Neurobiol Lab, NCIC Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| |
Collapse
|
8
|
Kammoun M, Pouletaut P, Morandat S, Subramaniam M, Hawse JR, Bensamoun SF. Krüppel-like factor 10 regulates the contractile properties of skeletal muscle fibers in mice. Muscle Nerve 2021; 64:765-769. [PMID: 34486132 DOI: 10.1002/mus.27412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 11/07/2022]
Abstract
INTRODUCTION/AIMS Klf10 is a member of the Krüppel-like family of transcription factors, which is implicated in mediating muscle structure (fiber size, organization of the sarcomere), muscle metabolic activity (respiratory chain), and passive force. The aim of this study was to further characterize the roles of Klf10 in the contractile properties of skeletal muscle fibers. METHODS Fifty-two single fibers were extracted from female wild-type (WT) and Klf10 knockout (KO) oxidative (soleus) and glycolytic (extensor digitorum longus [EDL]) skinned muscles. Each fiber was immersed successively in relaxing (R), washing (W), and activating (A) solutions. Calcium was included in the activating solution to induce a maximum contraction of the fiber. The maximum force (Fmax ) was measured and normalized to the cross-sectional area to obtain the maximum stress (Stressmax ). After a steady state in contraction was reached, a quick stretch-release was performed; the force at the maximum stretch (Fstretch ) was measured and the stiffness was assessed. RESULTS Deletion of the Klf10 gene induced changes in the contractile parameters (Fmax , Stressmax , Stiffness), which were lower and higher for soleus and EDL fibers compared with littermates, respectively. These measurements also revealed changes in the proportion and resistance of attached cross-bridges. DISCUSSION Klf10 plays a major role in the homeostasis of the contractile behavior of skeletal muscle fibers in a muscle fiber type-specific manner. These findings further implicate important roles for Klf10 in skeletal muscle function and shed new light on understanding the molecular processes regulating the contractility of skeletal muscle fibers.
Collapse
Affiliation(s)
- Malek Kammoun
- Biomechanics and Bioengineering, Centre de recherche Royallieu, Université de technologie de Compiègne, Compiègne Cedex, France
| | - Philippe Pouletaut
- Biomechanics and Bioengineering, Centre de recherche Royallieu, Université de technologie de Compiègne, Compiègne Cedex, France
| | - Sandrine Morandat
- Biomechanics and Bioengineering, Centre de recherche Royallieu, Université de technologie de Compiègne, Compiègne Cedex, France
| | - Malayannan Subramaniam
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sabine F Bensamoun
- Biomechanics and Bioengineering, Centre de recherche Royallieu, Université de technologie de Compiègne, Compiègne Cedex, France
| |
Collapse
|
9
|
Zou Z, Chai M, Guo F, Fu X, Lan Y, Cao S, Liu J, Tian L, An G. MicroRNA-126 engineered muscle-derived stem cells attenuates cavernosa injury-induced erectile dysfunction in rats. Aging (Albany NY) 2021; 13:14399-14415. [PMID: 34031263 PMCID: PMC8202866 DOI: 10.18632/aging.203057] [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: 07/22/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cavernosa injury is a common cause of organic erectile dysfunction (ED), which requires safe and effective treatments. In the present study, the therapeutic efficiency of muscle-derived stem cells (MDSCs) modified with microRNA-126 (miR-126) was determined in rats with cavernosa injury. METHODS MDSCs were transfected with miR-126 and then were transplanted into rats with cavernosa injury. Erectile function, vascular function (western blot and immunofluorescence), extraction, and detection of exosomes were then undertaken. RESULTS On the 28th day after transplantation, the highest value of intra-cavernous pressure (ICP)/mean arterial pressure (MAP) in rats of miRNA-126 group (0.84 ± 0.14) was observed (Control: 0.38 ± 0.07; MDSC: 0.54 ± 0.11, Vector: 0.60 ± 0.02; respectively). Treatment of miRNA-126-modified-MDSCs remarkably strengthened vascular structure, supported by hematoxylin-eosin staining. The expression of CD31, von Willebrand Factor and vascular endothelial factors were higher than those in other groups, indicating improved vascular function. In vitro mechanism studies showed that exosomes containing miR-126 isolated from MDSCs promoted angiogenesis and attenuated apoptosis of human umbilical venous endothelial cells. Finally, insulin receptor substrate 1 and Krüppel-like factor 10 were determined as the direct target genes of miR-126. CONCLUSIONS MiR-126 engineered MDSCs notably repaired cavernosa injury in rats via vascular reconstruction by directly targeting IRS1 and KLF10, in which the exosomes secreted by MDSCs played a critical role.
Collapse
Affiliation(s)
- Zihao Zou
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Muyuan Chai
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, PR China
| | - Feixiang Guo
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Fu
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Yu Lan
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Shuqi Cao
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Jianan Liu
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Long Tian
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Geng An
- Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| |
Collapse
|
10
|
You M, Zhang L, Zhang X, Fu Y, Dong X. MicroRNA-197-3p Inhibits the Osteogenic Differentiation in Osteoporosis by Down-Regulating KLF 10. Clin Interv Aging 2021; 16:107-117. [PMID: 33469278 PMCID: PMC7810594 DOI: 10.2147/cia.s269171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022] Open
Abstract
Background Studies have shown that microRNA (miRNA) regulates gene expression of osteoporosis (OS). It is known that miR-197-3p is abnormally expressed in osteoporosis. This study is to investigate the mechanism of miR-197-3p in regulating osteoblast differentiation. Methods Rats were ovariectomized to establish an animal model of postmenopausal osteoporosis. The expression of miR-197-3p and KLF10 was detected in ovariectomized rat models. Primary osteoblasts and MC3T-E1 cells were divided into the control group, miR-197-3p inhibitor group, NC inhibitor group and miR-197-3p inhibitor + si-KLF10 group. The expression of miR-197-3p and Kruppel-like factor 10 (KLF10) was detected by qRT-PCR and Western blot. The relationship between miR-197-3p and KLF10 was analyzed by bioinformatics and luciferase reporter assay. Cell viability was evaluated by MTT assay. The ALP activity measurement and mineralization analysis were performed. Results The expression of miR-197-3p was significantly raised in ovariectomized osteoporosis rats. During the differentiation of osteoblasts, the expression of miR-197-3p was significantly decreased, while the expression of KLF10 was significantly raised in primary osteoblasts and MC3E3T1 cells. The expression of RUNX2, ALP, OCN and OSX in miR-197-3p inhibitor group and MC3T3-E1 group was significantly raised, and the cell survival rate and mineralized nodule were raised as well. KLF10 may be the downstream target gene of miR-197-3p. After co-transfection of miR-197-3p inhibitor and si-klf10, ALP, Runx2, OCN and OSX mRNA, cell survival rate and mineralized nodule were significantly decreased in primary osteoblasts and MC3T3-E1 cells. Conclusion MiR-197-3p Inhibition promoted osteoblast differentiation and reduced OS by up-regulating KLF10.
Collapse
Affiliation(s)
- Murong You
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Liang Zhang
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Xiaoxiang Zhang
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Yang Fu
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Xieping Dong
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| |
Collapse
|
11
|
Lee J, Oh AR, Lee HY, Moon YA, Lee HJ, Cha JY. Deletion of KLF10 Leads to Stress-Induced Liver Fibrosis upon High Sucrose Feeding. Int J Mol Sci 2020; 22:ijms22010331. [PMID: 33396939 PMCID: PMC7794950 DOI: 10.3390/ijms22010331] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a consequence of chronic liver injury associated with chronic viral infection, alcohol abuse, and nonalcoholic fatty liver. The evidence from clinical and animal studies indicates that transforming growth factor-β (TGF-β) signaling is associated with the development of liver fibrosis. Krüppel-like factor 10 (KLF10) is a transcription factor that plays a significant role in TGF-β-mediated cell growth, apoptosis, and differentiation. In recent studies, it has been reported to be associated with glucose homeostasis and insulin resistance. In the present study, we investigated the role of KLF10 in the progression of liver disease upon a high-sucrose diet (HSD) in mice. Wild type (WT) and Klf10 knockout (KO) mice were fed either a control chow diet or HSD (50% sucrose) for eight weeks. Klf10 KO mice exhibited significant hepatic steatosis, inflammation, and liver injury upon HSD feeding, whereas the WT mice exhibited mild hepatic steatosis with no apparent liver injury. The livers of HSD-fed Klf10 KO mice demonstrated significantly increased endoplasmic reticulum stress, oxidative stress, and proinflammatory cytokines. Klf10 deletion led to the development of sucrose-induced hepatocyte cell death both in vivo and in vitro. Moreover, it significantly increased fibrogenic gene expression and collagen accumulation in the liver. Increased liver fibrosis was accompanied by increased phosphorylation and nuclear localization of Smad3. Here, we demonstrate that HSD-fed mice develop a severe liver injury in the absence of KLF10 due to the hyperactivation of the endoplasmic reticulum stress response and CCAAT/enhance-binding protein homologous protein (CHOP)-mediated apoptosis of hepatocytes. The current study suggests that KLF10 plays a protective role against the progression of hepatic steatosis into liver fibrosis in a lipogenic state.
Collapse
Affiliation(s)
- Junghoon Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Ah-Reum Oh
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Hui-Young Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University School of Medicine, Incheon 22212, Korea;
| | - Ho-Jae Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Correspondence: (H.-J.L.); (J.-Y.C.); Tel.: +82-32-899-6054 (H.-J.L.); +82-32-899-6070 (J.-Y.C.)
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea; (J.L.); (A.-R.O.); (H.-Y.L.)
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon 21565, Korea
- Correspondence: (H.-J.L.); (J.-Y.C.); Tel.: +82-32-899-6054 (H.-J.L.); +82-32-899-6070 (J.-Y.C.)
| |
Collapse
|
12
|
Kobayashi S, Nagafuchi Y, Okubo M, Sugimori Y, Shirai H, Hatano H, Junko M, Yanaoka H, Takeshima Y, Ota M, Iwasaki Y, Sumitomo S, Okamura T, Yamamoto K, Shoda H, Fujio K. Integrated bulk and single-cell RNA-sequencing identified disease-relevant monocytes and a gene network module underlying systemic sclerosis. J Autoimmun 2020; 116:102547. [PMID: 33039247 DOI: 10.1016/j.jaut.2020.102547] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Immunological disturbances have been reported in systemic sclerosis (SSc). This study assessed the transcriptome disturbances in immune cell subsets in SSc and characterized a disease-related gene network module and immune cell cluster at single cell resolution. METHODS Twenty-one Japanese SSc patients were enrolled and compared with 13 age- and sex-matched healthy controls (HC). Nineteen peripheral blood immune cell subsets were sorted by flow cytometry and bulk RNA-seq analysis was performed for each. Differential expression and pathway analyses were conducted. Iterative weighted gene correlation network analysis (iWGCNA) of each subset revealed clustered co-expressed gene network modules. Random forest analysis prioritized a disease-related gene module. Single cell RNA-seq analysis of 878 monocytes was integrated with bulk RNA-seq analysis and with a public database for single cell RNA-seq analysis of SSc patients. RESULTS Inflammatory pathway genes were differentially expressed in widespread immune cell subsets of SSc. An inflammatory gene module from CD16+ monocytes, which included KLF10, PLAUR, JUNB and JUND, showed the greatest discrimination between SSc and HC. One of the clusters of SSc monocytes identified by single-cell RNA-seq analysis characteristically expressed these inflammatory co-expressed genes and was similar to lung infiltrating FCN1hi monocytes expressing IL1B. CONCLUSIONS Our integrated analysis of bulk and single cell RNA-seq analysis identified an inflammatory gene module and a cluster of monocytes that are relevant to SSc pathophysiology. They could serve as candidate novel therapeutic targets in SSc.
Collapse
Affiliation(s)
- Satomi Kobayashi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Mai Okubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yusuke Sugimori
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Harumi Shirai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Hiroaki Hatano
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Maeda Junko
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Haruyuki Yanaoka
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Immuno-Rheumatology Center, St. Luke's International Hospital, St. Luke's International University, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan.
| | - Yusuke Takeshima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
13
|
Alcalde-Estévez E, Asenjo-Bueno A, Sosa P, Olmos G, Plaza P, Caballero-Mora MÁ, Rodríguez-Puyol D, Ruíz-Torres MP, López-Ongil S. Endothelin-1 induces cellular senescence and fibrosis in cultured myoblasts. A potential mechanism of aging-related sarcopenia. Aging (Albany NY) 2020; 12:11200-11223. [PMID: 32572011 PMCID: PMC7343454 DOI: 10.18632/aging.103450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Endothelial dysfunction, with increased endothelin-1 (ET-1) synthesis, and sarcopenia, characterized by the loss of muscular mass and strength, are two aging–related conditions. However, a relationship between them has not been already established. The aim of this study was to determine whether ET-1 induces senescence and fibrosis in cultured murine myoblasts, which could be involved in the development of sarcopenia related to aging. For this purpose, myoblasts were incubated with ET-1 to assess cellular senescence, analyzed by senescence associated β-galactosidase activity and p16 expression; and fibrosis, assessed by fibronectin expression. ET-1 induced myoblast senescence and fibrosis through ETA receptor. The use of antioxidants and several antagonists revealed that ET-1 effect on senescence and fibrosis depended on ROS production and activation of PI3K-AKT-GSK pathway. To stress the in vivo relevance of these results, circulating ET-1, muscular strength, muscular fibrosis and p16 expression were measured in male C57Bl6 mice from 5-18-24-months-old. Old mice shown high levels of ET-1 correlated with muscular fibrosis, muscular p16 expression and loss of muscle strength. In conclusion, ET-1 promotes fibrosis and senescence in cultured myoblasts, similar results were found in old mice, suggesting a potential role for ET-1 in the development of sarcopenia related to aging.
Collapse
Affiliation(s)
- Elena Alcalde-Estévez
- Departamento Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid 28871, Spain
| | - Ana Asenjo-Bueno
- Unidad de Investigación de la Fundación para la Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid 28805, Spain
| | - Patricia Sosa
- Departamento Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid 28871, Spain
| | - Gemma Olmos
- Departamento Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid 28871, Spain.,Instituto Reina Sofía de Investigación Nefrológica (IRSIN) de la Fundación Renal Iñigo Álvarez de Toledo (FRIAT), Madrid 28003, Spain.,Area 3-Fisiología y Fisiopatología Renal y Vascular del IRYCIS, Madrid 28046, Spain
| | - Patricia Plaza
- Unidad de Investigación de la Fundación para la Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid 28805, Spain
| | | | - Diego Rodríguez-Puyol
- Instituto Reina Sofía de Investigación Nefrológica (IRSIN) de la Fundación Renal Iñigo Álvarez de Toledo (FRIAT), Madrid 28003, Spain.,Area 3-Fisiología y Fisiopatología Renal y Vascular del IRYCIS, Madrid 28046, Spain.,Servicio de Nefrología del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid 28805, Spain
| | - María Piedad Ruíz-Torres
- Departamento Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid 28871, Spain.,Instituto Reina Sofía de Investigación Nefrológica (IRSIN) de la Fundación Renal Iñigo Álvarez de Toledo (FRIAT), Madrid 28003, Spain.,Area 3-Fisiología y Fisiopatología Renal y Vascular del IRYCIS, Madrid 28046, Spain
| | - Susana López-Ongil
- Unidad de Investigación de la Fundación para la Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid 28805, Spain.,Instituto Reina Sofía de Investigación Nefrológica (IRSIN) de la Fundación Renal Iñigo Álvarez de Toledo (FRIAT), Madrid 28003, Spain.,Area 3-Fisiología y Fisiopatología Renal y Vascular del IRYCIS, Madrid 28046, Spain
| |
Collapse
|
14
|
Yang P, Aweya JJ, Yao D, Wang F, Lun J, Hong Y, Sun K, Zhang Y. The krüppel-like factor of Penaeus vannamei negatively regulates transcription of the small subunit hemocyanin gene as part of shrimp immune response. FISH & SHELLFISH IMMUNOLOGY 2020; 100:397-406. [PMID: 32201349 DOI: 10.1016/j.fsi.2020.03.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/10/2020] [Accepted: 03/17/2020] [Indexed: 06/10/2023]
Abstract
Hemocyanin is a multifunctional respiratory glycoprotein, which has also been implicated in other biological functions in shrimp. Moreover, recent studies have revealed that hemocyanin is also involved in a broad range of immune-related activities in shrimp. However, in spite of the considerable interest in unraveling the reasons behind the multiple immune-related functions of hemocyanin, little is known about its transcriptional regulation. Here, DNA pull-down and Liquid Chromatography - Tandem Mass Spectrometry (LC-MS/MS) analyses were used to isolate and identify the putative transcription factor(s) that are involved in the transcriptional regulation of the small subunit hemocyanin gene of Penaeus vannamei (PvHMCs). Krüppel-like factor (designated PvKruppel), a zinc finger transcription factor homolog in P. vannamei, was identified among the putative transcription factors, while bioinformatics analysis revealed the presence of Krüppel-like factor binding site (KLF motif) on the core promoter region of PvHMCs. Mutational analysis and electrophoretic mobility shift assay (EMSA) confirmed that PvKruppel could bind to the KLF motif on the core promoter region of PvHMCs. Moreover, in response to lipopolysaccharide (LPS), Vibrio parahaemolyticus and white spot syndrome virus (WSSV) challenge, transcript levels of PvKruppel and PvHMCs were negatively correlated. Furthermore, overexpression of PvKruppel significantly reduced the promoter activity of PvHMCs, while PvKruppel knockdown by RNA interference or lipopolysaccharides (LPS) stimulation resulted in a significant increase in the transcript level of PvHMCs. Taken together, our present study provides mechanistic insights into the transcriptional regulation of PvHMCs by PvKruppel during shrimp immune response to pathogens.
Collapse
Affiliation(s)
- Peikui Yang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Jude Juventus Aweya
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Defu Yao
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Fan Wang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Jingsheng Lun
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Yujian Hong
- Guangdong Yuequn Marine Biological Research and Development Co., Ltd., Jieyang, 515200, China
| | - Kaihui Sun
- Guangdong Yuequn Marine Biological Research and Development Co., Ltd., Jieyang, 515200, China
| | - Yueling Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China.
| |
Collapse
|
15
|
Kammoun M, Piquereau J, Nadal‐Desbarats L, Même S, Beuvin M, Bonne G, Veksler V, Le Fur Y, Pouletaut P, Même W, Szeremeta F, Constans J, Bruinsma ES, Nelson Holte MH, Najafova Z, Johnsen SA, Subramaniam M, Hawse JR, Bensamoun SF. Novel role of Tieg1 in muscle metabolism and mitochondrial oxidative capacities. Acta Physiol (Oxf) 2020; 228:e13394. [PMID: 31560161 DOI: 10.1111/apha.13394] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/26/2022]
Abstract
AIM Tieg1 is involved in multiple signalling pathways, human diseases, and is highly expressed in muscle where its functions are poorly understood. METHODS We have utilized Tieg1 knockout (KO) mice to identify novel and important roles for this transcription factor in regulating muscle ultrastructure, metabolism and mitochondrial functions in the soleus and extensor digitorum longus (EDL) muscles. RNA sequencing, immunoblotting, transmission electron microscopy, MRI, NMR, histochemical and mitochondrial function assays were performed. RESULTS Loss of Tieg1 expression resulted in altered sarcomere organization and a significant decrease in mitochondrial number. Histochemical analyses demonstrated an absence of succinate dehydrogenase staining and a decrease in cytochrome c oxidase (COX) enzyme activity in KO soleus with similar, but diminished, effects in the EDL. Decreased complex I, COX and citrate synthase (CS) activities were detected in the soleus muscle of KO mice indicating altered mitochondrial function. Complex I activity was also diminished in KO EDL. Significant decreases in CS and respiratory chain complex activities were identified in KO soleus. 1 H-NMR spectra revealed no significant metabolic difference between wild-type and KO muscles. However, 31 P spectra revealed a significant decrease in phosphocreatine and ATPγ. Altered expression of 279 genes, many of which play roles in mitochondrial and muscle function, were identified in KO soleus muscle. Ultimately, all of these changes resulted in an exercise intolerance phenotype in Tieg1 KO mice. CONCLUSION Our findings have implicated novel roles for Tieg1 in muscle including regulation of gene expression, metabolic activity and organization of tissue ultrastructure. This muscle phenotype resembles diseases associated with exercise intolerance and myopathies of unknown consequence.
Collapse
Affiliation(s)
- Malek Kammoun
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| | - Jerome Piquereau
- Signalling and Cardiovascular Pathophysiology ‐ UMR‐S 1180 Université Paris‐Sud INSERM Université Paris‐Saclay Châtenay‐Malabry France
| | | | - Sandra Même
- CNRS UPR4301 Centre de Biophysique Moléculaire Orléans France
| | - Maud Beuvin
- Inserm U974 Centre de Recherche en Myologie Sorbonne Université Paris France
| | - Gisèle Bonne
- Inserm U974 Centre de Recherche en Myologie Sorbonne Université Paris France
| | - Vladimir Veksler
- Signalling and Cardiovascular Pathophysiology ‐ UMR‐S 1180 Université Paris‐Sud INSERM Université Paris‐Saclay Châtenay‐Malabry France
| | - Yann Le Fur
- Aix‐Marseille University CNRS CRMBM Marseille France
| | - Philippe Pouletaut
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| | - William Même
- CNRS UPR4301 Centre de Biophysique Moléculaire Orléans France
| | | | - Jean‐Marc Constans
- Institut Faire Faces EA Chimère Imagerie et Radiologie Médicale CHU Amiens Amiens France
| | | | | | - Zeynab Najafova
- Department of General, Visceral and Pediatric Surgery University Medical Center Göttingen Göttingen Germany
| | - Steven A. Johnsen
- Department of General, Visceral and Pediatric Surgery University Medical Center Göttingen Göttingen Germany
| | | | - John R. Hawse
- Department of Biochemistry and Molecular Biology Mayo Clinic Rochester MN USA
| | - Sabine F. Bensamoun
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| |
Collapse
|
16
|
GPCR-induced YAP activation sensitizes fibroblasts to profibrotic activity of TGFβ1. PLoS One 2020; 15:e0228195. [PMID: 32053631 PMCID: PMC7018035 DOI: 10.1371/journal.pone.0228195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/09/2020] [Indexed: 12/16/2022] Open
Abstract
Tissue fibrosis is a pathological condition characterized by uncontrolled fibroblast activation that ultimately leads to organ failure. The TGFβ1 pathway, one of the major players in establishment of the disease phenotype, is dependent on the transcriptional co-activators YAP/TAZ. We were interested whether fibroblasts can be sensitized to TGFβ1 by activation of the GPCR/YAP/TAZ axis and whether this mechanism explains the profibrotic properties of diverse GPCR ligands. We found that LPA, S1P and thrombin cooperate in human dermal fibroblasts with TGFβ1 to induce extracellular matrix synthesis, myofibroblast marker expression and cytokine secretion. Whole genome expression profiling identified a YAP/TAZ signature behind the synergistic profibrotic effects of LPA and TGFβ1. LPA, S1P and thrombin stimulation led to activation of the Rho-YAP axis, an increase of nuclear YAP-Smad2 complexes and enhanced expression of profibrotic YAP/Smad2-target genes. More generally, dermal, cardiac and lung fibroblast responses to TGFβ1 could be enhanced by increasing YAP nuclear levels (with GPCR ligands LPA, S1P, thrombin or Rho activator) and inhibited by decreasing nuclear YAP (with Rho inhibitor, forskolin, latrunculin B or 2-deoxy-glucose). Thus, we present here a conceptually interesting finding that fibroblast responses to TGFβ1 can be predicted based on the nuclear levels of YAP and modulated by stimuli/treatments that change YAP nuclear levels. Our study contributes to better understanding of fibrosis as a complex interplay of signalling pathways and proposes YAP/TAZ as promising targets in the treatment of fibrosis.
Collapse
|
17
|
Identification of qPCR reference genes suitable for normalizing gene expression in the mdx mouse model of Duchenne muscular dystrophy. PLoS One 2019; 14:e0211384. [PMID: 30699165 PMCID: PMC6353192 DOI: 10.1371/journal.pone.0211384] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
The mdx mouse is the most widely-used animal model of the human disease Duchenne muscular dystrophy, and quantitative PCR analysis of gene expression in the muscles of this animal plays a key role in the study of pathogenesis and disease progression and in evaluation of potential therapeutic interventions. Normalization to appropriate stably-expressed reference genes is essential for accurate quantitative measurement, but determination of such genes is challenging: healthy and dystrophic muscles present very different transcriptional environments, further altering with disease progression and muscle use, raising the possibility that no single gene or combination of genes may be stable under all experimental comparative scenarios. Despite the pedigree of this animal model, this problem remains unaddressed. The aim of this work was therefore to comprehensively assess reference gene suitability in the muscles of healthy and dystrophic mice, identifying reference genes appropriate for specific experimental comparisons, and determining whether an essentially universally-applicable set of genes exists. Using a large sample collection comprising multiple muscles (including the tibialis anterior, diaphragm and heart muscles) taken from healthy and mdx mice at three disease-relevant ages, and a panel of sixteen candidate reference genes (FBXO38, FBXW2, MON2, ZFP91, HTATSF1, GAPDH, ACTB, 18S, CDC40, SDHA, RPL13a, CSNK2A2, AP3D1, PAK1IP1, B2M and HPRT1), we used the geNorm, BestKeeper and Normfinder algorithms to identify genes that were stable under multiple possible comparative scenarios. We reveal that no single gene is stable under all conditions, but a normalization factor derived from multiple genes (RPL13a, CSNK2A2, AP3D1 and the widely-used ACTB) appears suitable for normalizing gene expression in both healthy and dystrophic mouse muscle regardless of muscle type or animal age. We further show that other popular reference genes, including GAPDH, are markedly disease- or muscle-type correlated. This study demonstrates the importance of empirical reference gene identification, and should serve as a valuable resource for investigators wishing to study gene expression in mdx mice.
Collapse
|
18
|
Rane MJ, Zhao Y, Cai L. Krϋppel-like factors (KLFs) in renal physiology and disease. EBioMedicine 2019; 40:743-750. [PMID: 30662001 PMCID: PMC6414320 DOI: 10.1016/j.ebiom.2019.01.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
Dysregulated Krϋppel-like factor (KLF) gene expression appears in many disease-associated pathologies. In this review, we discuss physiological functions of KLFs in the kidney with a focus on potential pharmacological modulation/therapeutic applications of these KLF proteins. KLF2 is critical to maintaining endothelial barrier integrity and preventing gap formations and in prevention of glomerular endothelial cell and podocyte damage in diabetic mice. KLF4 is renoprotective in the setting of AKI and is a critical regulator of proteinuria in mice and humans. KLF6 expression in podocytes preserves mitochondrial function and prevents podocyte apoptosis, while KLF5 expression prevents podocyte apoptosis by blockade of ERK/p38 MAPK pathways. KLF15 is a critical regulator of podocyte differentiation and is protective against podocyte injury. Loss of KLF4 and KLF15 promotes renal fibrosis, while fibrotic kidneys have increased KLF5 and KLF6 expression. For therapeutic modulation of KLFs, continued screening of small molecules will promote drug discoveries targeting KLF proteins.
Collapse
Affiliation(s)
- Madhavi J Rane
- Department of Medicine, Division Nephrology, Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40292, USA.
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
19
|
Tang P, Zhu R, Gu Y, Gu JM, Xie ZA, Xu JQ, Zhao XD, Huang KM, Wang JY, Jiang XS, Fan SW, Hu ZJ. Ascorbic Acid Attenuates Multifidus Muscles Injury and Atrophy After Posterior Lumbar Spine Surgery by Suppressing Inflammation and Oxidative Stress in a Rat Model. Spine (Phila Pa 1976) 2018; 43:E1249-E1259. [PMID: 29649092 DOI: 10.1097/brs.0000000000002669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN A rat model of multifidus muscles injury and atrophy after posterior lumbar spine surgery. OBJECTIVE We determined the effect of ascorbic acid (AA) on the postoperative multifidus muscles in rat model. SUMMARY OF BACKGROUND DATA Previous studies show oxidative stress and inflammation are two main molecular mechanisms in multifidus muscle injury and atrophy after posterior lumbar surgery. AA may have a protective effect in postoperative multifidus muscles. METHODS Rats were divided into sham surgery, control surgery, and surgery plus AA groups. Multifidus muscles of the control and AA groups were excised from the osseous structures. The muscles were retracted continuously for 2 hours. In the sham and AA groups, AA was administered via oral gavage daily in the first week. In each group, the oxidative stress was evaluated by measuring malondialdehyde (MDA) and Total superoxide dismutase (T-SOD). The inflammation, fat degeneration, or fibrosis of multifidus muscle were evaluated by quantitative real-time polymerase chain reaction (q-PCR), histology, or immunohistochemical analysis. RESULTS T-SOD activity was significantly lower in the control group than that in the AA group in the first week. MDA levels were significantly higher in the AA group. Interleukin-6 and tumor necrosis factor-α in multifidus muscles also showed significant differences when treated with AA. The inflammation score on histology was significantly lower in the AA group postoperatively in the first week. In the long run, marker genes for fibrosis and fat degeneration, and fibrosis and fat degeneration scores, were significantly lower in the AA than the control group on days 14 and 28 postoperatively. CONCLUSION In conclusion, AA attenuated the oxidative stress and inflammation response in the postoperative multifidus muscles, and remarkable differences were observed from the histological assessment and related marker genes expression. Our results provided important insight into the anti-inflammatory and anti-oxidative effects of AA in the postoperative multifidus muscles. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Pan Tang
- Department of Orthopaedics, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China.,Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ren Zhu
- Department of Orthopaedics, Yiwu Chowzhou Hospital, Yiwu, China
| | - Yu Gu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Ming Gu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zi-Ang Xie
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Qi Xu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang-De Zhao
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kang-Mao Huang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ji-Ying Wang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue-Sheng Jiang
- Department of Orthopaedics, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Shun-Wu Fan
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Jun Hu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|