1
|
Yamauchi N, Ashida Y, Naito A, Tokuda N, Niibori A, Motohashi N, Aoki Y, Yamada T. Fatigue Resistance and Mitochondrial Adaptations to Isometric Interval Training in Dystrophin-Deficient Muscle: Role of Contractile Load. FASEB J 2025; 39:e70631. [PMID: 40366239 PMCID: PMC12077386 DOI: 10.1096/fj.202500618rr] [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: 03/13/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
In normal mouse skeletal muscles, interval training (IT)-mimicking neuromuscular electrical stimulation enhances muscle fatigue resistance and mitochondrial content, with greater gains observed at high (100 Hz stimulation, IT100) compared to low (20 Hz stimulation, IT20) contractile load. In this study, we compared the effects of repeated IT100 and IT20 on fatigue resistance and mitochondrial adaptations in young male mdx52 mice (4- to 6-week-old), an animal model for Duchenne muscular dystrophy. Plantar flexor muscles were stimulated in vivo using supramaximal electrical stimulation to induce isometric contractions every other day for 4 weeks (a total of 15 sessions). In non-trained muscles of mdx52 mice, decreased fatigue resistance was associated with reduced citrate synthase activity, lower peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC-1α) protein expression, and diminished levels of mitochondrial respiratory chain complex II, and an increased percentage of Evans Blue dye-positive areas. IT100, but not IT20, markedly improved fatigue resistance and restored all these alterations in mdx52 mice. Furthermore, an acute session of IT100, but not IT20, led to increased phosphorylation of p38 mitogen-activated protein kinase (MAPK) and elevated mRNA levels of PGC-1α, which were blocked by the p38 MAPK inhibitor SB203580. These findings suggest that contractile load is a key determinant of isometric IT-induced improvements in fatigue resistance, even in dystrophin-deficient muscles, potentially through a p38 MAPK/PGC-1α-mediated increase in mitochondrial content.
Collapse
Affiliation(s)
- Nao Yamauchi
- Graduate School of Health SciencesSapporo Medical UniversitySapporoJapan
- The Japan Society for the Promotion of Science (JSPS)TokyoJapan
| | - Yuki Ashida
- The Japan Society for the Promotion of Science (JSPS)TokyoJapan
- Department of Molecular TherapyNational Institute of Neuroscience, National Center of Neurology and PsychiatryTokyoJapan
| | - Azuma Naito
- Graduate School of Health SciencesSapporo Medical UniversitySapporoJapan
| | - Nao Tokuda
- Graduate School of Health SciencesSapporo Medical UniversitySapporoJapan
- The Japan Society for the Promotion of Science (JSPS)TokyoJapan
| | - Ayaka Niibori
- Graduate School of Health SciencesSapporo Medical UniversitySapporoJapan
| | - Norio Motohashi
- Department of Molecular TherapyNational Institute of Neuroscience, National Center of Neurology and PsychiatryTokyoJapan
| | - Yoshitsugu Aoki
- Department of Molecular TherapyNational Institute of Neuroscience, National Center of Neurology and PsychiatryTokyoJapan
| | - Takashi Yamada
- Graduate School of Health SciencesSapporo Medical UniversitySapporoJapan
- Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| |
Collapse
|
2
|
Baum O. Expression of neuronal NO synthase α- and β-isoforms in skeletal muscle of mice. Biochem J 2024; 481:601-613. [PMID: 38592741 DOI: 10.1042/bcj20230458] [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: 11/02/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/10/2024]
Abstract
Knowledge of the primary structure of neuronal NO synthase (nNOS) in skeletal muscle is still conflicting and needs further clarification. To elucidate the expression patterns of nNOS isoforms at both mRNA and protein level, systematic reverse transcription (RT)-PCR and epitope mapping by qualitative immunoblot analysis on skeletal muscle of C57/BL6 mice were performed. The ability of the nNOS isoforms to form aggregates was characterized by native low-temperature polyacrylamide electrophoresis (LT-PAGE). The molecular analysis was focused on the rectus femoris (RF) muscle, a skeletal muscle with a nearly balanced ratio of nNOS α- and β-isoforms. RT-PCR amplificates from RF muscles showed exclusive exon-1d mRNA expression, either with or without exon-μ. Epitope mapping demonstrated the simultaneous expression of the nNOS splice variants α/μ, α/non-μ, β/μ and β/non-μ. Furthermore, immunoblotting suggests that the transition between nNOS α- and β-isoforms lies within exon-3. In LT-PAGE, three protein nNOS associated aggregates were detected in homogenates of RF muscle and tibialis anterior muscle: a 320 kDa band containing nNOS α-isoforms, while 250 and 300 kDa bands consist of nNOS β-isoforms that form homodimers or heterodimers with non-nNOS proteins.
Collapse
Affiliation(s)
- Oliver Baum
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| |
Collapse
|
3
|
Yamauchi N, Tamai K, Kimura I, Naito A, Tokuda N, Ashida Y, Motohashi N, Aoki Y, Yamada T. High-intensity interval training in the form of isometric contraction improves fatigue resistance in dystrophin-deficient muscle. J Physiol 2023; 601:2917-2933. [PMID: 37184335 DOI: 10.1113/jp284532] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023] Open
Abstract
Duchenne muscular dystrophy is a genetic muscle-wasting disorder characterized by progressive muscle weakness and easy fatigability. Here we examined whether high-intensity interval training (HIIT) in the form of isometric contraction improves fatigue resistance in skeletal muscle from dystrophin-deficient mdx52 mice. Isometric HIIT was performed on plantar flexor muscles in vivo with supramaximal electrical stimulation every other day for 4 weeks (a total of 15 sessions). In the non-trained contralateral gastrocnemius muscle from mdx52 mice, the decreased fatigue resistance was associated with a reduction in the amount of peroxisome proliferator-activated receptor γ coactivator 1-α, citrate synthase activity, mitochondrial respiratory complex II, LC3B-II/I ratio, and mitophagy-related gene expression (i.e. Pink1, parkin, Bnip3 and Bcl2l13) as well as an increase in the phosphorylation levels of Src Tyr416 and Akt Ser473, the amount of p62, and the percentage of Evans Blue dye-positive area. Isometric HIIT restored all these alterations and markedly improved fatigue resistance in mdx52 muscles. Moreover, an acute bout of HIIT increased the phosphorylation levels of AMP-activated protein kinase (AMPK) Thr172, acetyl CoA carboxylase Ser79, unc-51-like autophagy activating kinase 1 (Ulk1) Ser555, and dynamin-related protein 1 (Drp1) Ser616 in mdx52 muscles. Thus, our data show that HIIT with isometric contractions significantly mitigates histological signs of pathology and improves fatigue resistance in dystrophin-deficient muscles. These beneficial effects can be explained by the restoration of mitochondrial function via AMPK-dependent induction of the mitophagy programme and de novo mitochondrial biogenesis. KEY POINTS: Skeletal muscle fatigue is often associated with Duchenne muscular dystrophy (DMD) and leads to an inability to perform daily tasks, profoundly decreasing quality of life. We examined the effect of high-intensity interval training (HIIT) in the form of isometric contraction on fatigue resistance in skeletal muscle from the mdx52 mouse model of DMD. Isometric HIIT counteracted the reduced fatigue resistance as well as dystrophic changes in skeletal muscle of mdx52 mice. This beneficial effect could be explained by the restoration of mitochondrial function via AMP-activated protein kinase-dependent mitochondrial biogenesis and the induction of the mitophagy programme in the dystrophic muscles.
Collapse
Affiliation(s)
- Nao Yamauchi
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Katsuyuki Tamai
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Iori Kimura
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Azuma Naito
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Nao Tokuda
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Yuki Ashida
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
- The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
4
|
Baum O, Huber-Abel FAM, Flück M. nNOS Increases Fiber Type-Specific Angiogenesis in Skeletal Muscle of Mice in Response to Endurance Exercise. Int J Mol Sci 2023; 24:ijms24119341. [PMID: 37298293 DOI: 10.3390/ijms24119341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
We studied the relationship between neuronal NO synthase (nNOS) expression and capillarity in the tibialis anterior (TA) muscle of mice subjected to treadmill training. The mRNA (+131%) and protein (+63%) levels of nNOS were higher (p ≤ 0.05) in the TA muscle of C57BL/6 mice undergoing treadmill training for 28 days than in those of littermates remaining sedentary, indicating an up-regulation of nNOS by endurance exercise. Both TA muscles of 16 C57BL/6 mice were subjected to gene electroporation with either the pIRES2-ZsGreen1 plasmid (control plasmid) or the pIRES2-ZsGreen1-nNOS gene-inserted plasmid (nNOS plasmid). Subsequently, one group of mice (n = 8) underwent treadmill training for seven days, while the second group of mice (n = 8) remained sedentary. At study end, 12-18% of TA muscle fibers expressed the fluorescent reporter gene ZsGreen1. Immunofluorescence for nNOS was 23% higher (p ≤ 0.05) in ZsGreen1-positive fibers than ZsGreen1-negative fibers from the nNOS-transfected TA muscle of mice subjected to treadmill training. Capillary contacts around myosin heavy-chain (MHC)-IIb immunoreactive fibers (14.2%; p ≤ 0.05) were only higher in ZsGreen1-positive fibers than ZsGreen1-negative fibers in the nNOS-plasmid-transfected TA muscles of trained mice. Our observations are in line with an angiogenic effect of quantitative increases in nNOS expression, specifically in type-IIb muscle fibers after treadmill training.
Collapse
Affiliation(s)
- Oliver Baum
- Institute of Physiology, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | | | - Martin Flück
- Heart Repair and Regeneration Laboratory, Department EMC, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
5
|
Niba ETE, Awano H, Lee T, Takeshima Y, Shinohara M, Nishio H, Matsuo M. Dystrophin Dp71 Subisoforms Localize to the Mitochondria of Human Cells. Life (Basel) 2021; 11:life11090978. [PMID: 34575126 PMCID: PMC8468555 DOI: 10.3390/life11090978] [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: 06/21/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 11/26/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by deficiency in dystrophin, a protein product encoded by the DMD gene. Mitochondrial dysfunction is now attracting much attention as a central player in DMD pathology. However, dystrophin has never been explored in human mitochondria. Here, we analyzed dystrophin in cDNAs and mitochondrial fractions of human cells. Mitochondrial fraction was obtained using a magnetic-associated cell sorting (MACS) technology. Dystrophin was analyzed by reverse transcription (RT)-PCR and western blotting using an antibody against the dystrophin C-terminal. In isolated mitochondrial fraction from HEK293 cells, dystrophin was revealed as a band corresponding to Dp71b and Dp71ab subisoforms. Additionally, in mitochondria from HeLa, SH-SY5Y, CCL-136 and HepG2 cells, signals for Dp71b and Dp71ab were revealed as well. Concomitantly, dystrophin mRNAs encoding Dp71b and Dp71ab were disclosed by RT-PCR in these cells. Primary cultured myocytes from three dystrophinopathy patients showed various levels of mitochondrial Dp71 expression. Coherently, levels of mRNA were different in all cells reflecting the protein content, which indicated predominant accumulation of Dp71. Dystrophin was demonstrated to be localized to human mitochondrial fraction, specifically as Dp71 subisoforms. Myocytes derived from dystrophinopathy patients manifested different levels of mitochondrial Dp71, with higher expression revealed in myocytes from Becker muscular dystrophy (BMD) patient-derived myocytes.
Collapse
Affiliation(s)
- Emma Tabe Eko Niba
- Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
- Correspondence: ; Tel.: +81-78-382-5543
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Tomoko Lee
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Masakazu Shinohara
- Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Hisahide Nishio
- Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan;
| | - Masafumi Matsuo
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan;
| |
Collapse
|
6
|
Gao W, Huang M, Chen X, Chen J, Zou Z, Li L, Ji K, Nie Z, Yang B, Wei Z, Xu P, Jia J, Zhang Q, Shen H, Wang Q, Li K, Zhu L, Wang M, Ye S, Zeng S, Lin Y, Rong Z, Xu Y, Zhu P, Zhang H, Hao B, Liu Q. The role of S-nitrosylation of PFKM in regulation of glycolysis in ovarian cancer cells. Cell Death Dis 2021; 12:408. [PMID: 33859186 PMCID: PMC8050300 DOI: 10.1038/s41419-021-03681-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
One of the malignant transformation hallmarks is metabolism reprogramming, which plays a critical role in the biosynthetic needs of unchecked proliferation, abrogating cell death programs, and immunologic escape. However, the mechanism of the metabolic switch is not fully understood. Here, we found that the S-nitrosoproteomic profile of endogenous nitrogen oxide in ovarian cancer cells targeted multiple components in metabolism processes. Phosphofructokinase (PFKM), one of the most important regulatory enzymes of glycolysis, was S-nitrosylated by nitric oxide synthase NOS1 at Cys351. S-nitrosylation at Cys351 stabilized the tetramer of PFKM, leading to resist negative feedback of downstream metabolic intermediates. The PFKM-C351S mutation decreased the proliferation rate of cultured cancer cells, and reduced tumor growth and metastasis in the mouse xenograft model. These findings indicated that S-nitrosylation at Cys351 of PFKM by NOS1 contributes to the metabolic reprogramming of ovarian cancer cells, highlighting a critical role of endogenous nitrogen oxide on metabolism regulations in tumor progression.
Collapse
Affiliation(s)
- Wenwen Gao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xi Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhiwei Zou
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Southern Hospital Zengcheng Branch, Southern Medical University, Guangzhou, 528308, China
| | - Linlin Li
- First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
| | - Kaiyuan Ji
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhirui Nie
- Guangzhou Panyu Central Hospital, Guangzhou, 511400, China
| | - Bingsheng Yang
- Pearl River Hospital, Southern Medical University, Guangzhou, 528308, China
| | - Zibo Wei
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Pengfei Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junshuang Jia
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qianbing Zhang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongfen Shen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qianli Wang
- Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province, 450003, China
| | - Keyi Li
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lingqun Zhu
- Guangzhou Concord Cancer Center, Guangzhou, 528308, China
| | - Meng Wang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangyan Ye
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sisi Zeng
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying Lin
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhili Rong
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yang Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Peng Zhu
- Central Lab of Shenzhen Pingshan People's Hospital, Shenzhen, 518118, P. R. China
| | - Hui Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China.
- Platform of Metabolomics, Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China.
| | - Bingtao Hao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Genetic Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genoics, Henan Provincial People's Hospital Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China.
| | - Qiuzhen Liu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Pingshan General Hospital of Southern Medical University, Southern Medical University, Shenzhen, 518118, China.
| |
Collapse
|
7
|
Dowling P, Gargan S, Murphy S, Zweyer M, Sabir H, Swandulla D, Ohlendieck K. The Dystrophin Node as Integrator of Cytoskeletal Organization, Lateral Force Transmission, Fiber Stability and Cellular Signaling in Skeletal Muscle. Proteomes 2021; 9:9. [PMID: 33540575 PMCID: PMC7931087 DOI: 10.3390/proteomes9010009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
The systematic bioanalytical characterization of the protein product of the DMD gene, which is defective in the pediatric disorder Duchenne muscular dystrophy, led to the discovery of the membrane cytoskeletal protein dystrophin. Its full-length muscle isoform Dp427-M is tightly linked to a sarcolemma-associated complex consisting of dystroglycans, sarcoglyans, sarcospan, dystrobrevins and syntrophins. Besides these core members of the dystrophin-glycoprotein complex, the wider dystrophin-associated network includes key proteins belonging to the intracellular cytoskeleton and microtubular assembly, the basal lamina and extracellular matrix, various plasma membrane proteins and cytosolic components. Here, we review the central role of the dystrophin complex as a master node in muscle fibers that integrates cytoskeletal organization and cellular signaling at the muscle periphery, as well as providing sarcolemmal stabilization and contractile force transmission to the extracellular region. The combination of optimized tissue extraction, subcellular fractionation, advanced protein co-purification strategies, immunoprecipitation, liquid chromatography and two-dimensional gel electrophoresis with modern mass spectrometry-based proteomics has confirmed the composition of the core dystrophin complex at the sarcolemma membrane. Importantly, these biochemical and mass spectrometric surveys have identified additional members of the wider dystrophin network including biglycan, cavin, synemin, desmoglein, tubulin, plakoglobin, cytokeratin and a variety of signaling proteins and ion channels.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23F2H6 Maynooth, Co. Kildare, Ireland; (P.D.); (S.G.)
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23F2H6 Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23F2H6 Maynooth, Co. Kildare, Ireland; (P.D.); (S.G.)
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23F2H6 Maynooth, Co. Kildare, Ireland
| | - Sandra Murphy
- Newcastle Fibrosis Research Group, Newcastle University Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE24HH, UK;
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children’s Hospital, University of Bonn, D53113 Bonn, Germany; (M.Z.); (H.S.)
| | - Hemmen Sabir
- Department of Neonatology and Paediatric Intensive Care, Children’s Hospital, University of Bonn, D53113 Bonn, Germany; (M.Z.); (H.S.)
| | - Dieter Swandulla
- Institute of Physiology II, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23F2H6 Maynooth, Co. Kildare, Ireland; (P.D.); (S.G.)
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
8
|
Timpani CA, Mamchaoui K, Butler-Browne G, Rybalka E. Nitric Oxide (NO) and Duchenne Muscular Dystrophy: NO Way to Go? Antioxidants (Basel) 2020; 9:antiox9121268. [PMID: 33322149 PMCID: PMC7764682 DOI: 10.3390/antiox9121268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 01/09/2023] Open
Abstract
The discordance between pre-clinical success and clinical failure of treatment options for Duchenne Muscular Dystrophy (DMD) is significant. The termination of clinical trials investigating the phosphodiesterase inhibitors, sildenafil and tadalafil (which prolong the second messenger molecule of nitric oxide (NO) signaling), are prime examples of this. Both attenuated key dystrophic features in the mdx mouse model of DMD yet failed to modulate primary outcomes in clinical settings. We have previously attempted to modulate NO signaling via chronic nitrate supplementation of the mdx mouse but failed to demonstrate beneficial modulation of key dystrophic features (i.e., metabolism). Instead, we observed increased muscle damage and nitrosative stress which exacerbated MD. Here, we highlight that acute nitrite treatment of human DMD myoblasts is also detrimental and suggest strategies for moving forward with NO replacement therapy in DMD.
Collapse
Affiliation(s)
- Cara A. Timpani
- Institute for Health and Sport, Victoria University, Melbourne 8001, Victoria, Australia;
- Australian Institute for Musculoskeletal Science, St Albans 3021, Victoria, Australia
- Correspondence: ; Tel.: +61-3-8395-8206
| | - Kamel Mamchaoui
- Institut de Myologie, Sorbonne University, INSERM UMRS974 Paris, France; (K.M.); (G.B.-B.)
| | - Gillian Butler-Browne
- Institut de Myologie, Sorbonne University, INSERM UMRS974 Paris, France; (K.M.); (G.B.-B.)
| | - Emma Rybalka
- Institute for Health and Sport, Victoria University, Melbourne 8001, Victoria, Australia;
- Australian Institute for Musculoskeletal Science, St Albans 3021, Victoria, Australia
| |
Collapse
|
9
|
Zhao J, Yang HT, Wasala L, Zhang K, Yue Y, Duan D, Lai Y. Dystrophin R16/17 protein therapy restores sarcolemmal nNOS in trans and improves muscle perfusion and function. Mol Med 2019; 25:31. [PMID: 31266455 PMCID: PMC6607532 DOI: 10.1186/s10020-019-0101-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Background Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. Methods Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. Results Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. Conclusions Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton. Electronic supplementary material The online version of this article (10.1186/s10020-019-0101-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Hsiao Tung Yang
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
10
|
Patel A, Zhao J, Yue Y, Zhang K, Duan D, Lai Y. Dystrophin R16/17-syntrophin PDZ fusion protein restores sarcolemmal nNOSμ. Skelet Muscle 2018; 8:36. [PMID: 30466494 PMCID: PMC6251231 DOI: 10.1186/s13395-018-0182-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background Loss of sarcolemmal nNOSμ is a common manifestation in a wide variety of muscle diseases and contributes to the dysregulation of multiple muscle activities. Given the critical role sarcolemmal nNOSμ plays in muscle, restoration of sarcolemmal nNOSμ should be considered as an important therapeutic goal. Methods nNOSμ is anchored to the sarcolemma by dystrophin spectrin-like repeats 16 and 17 (R16/17) and the syntrophin PDZ domain (Syn PDZ). To develop a strategy that can independently restore sarcolemmal nNOSμ, we engineered an R16/17-Syn PDZ fusion construct and tested whether this construct alone is sufficient to anchor nNOSμ to the sarcolemma in three different mouse models of Duchenne muscular dystrophy (DMD). Results Membrane-associated nNOSμ is completely lost in DMD. Adeno-associated virus (AAV)-mediated delivery of the R16/17-Syn PDZ fusion construct successfully restored sarcolemmal nNOSμ in all three models. Further, nNOS restoration was independent of the dystrophin-associated protein complex. Conclusions Our results suggest that the R16/17-Syn PDZ fusion construct is sufficient to restore sarcolemmal nNOSμ in the dystrophin-null muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0182-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
11
|
Abstract
The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
12
|
Dombernowsky NW, Ölmestig JNE, Witting N, Kruuse C. Role of neuronal nitric oxide synthase (nNOS) in Duchenne and Becker muscular dystrophies - Still a possible treatment modality? Neuromuscul Disord 2018; 28:914-926. [PMID: 30352768 DOI: 10.1016/j.nmd.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) is involved in nitric oxide (NO) production and suggested to play a crucial role in blood flow regulation of skeletal muscle. During activation of the muscle, NO helps attenuate the sympathetic vasoconstriction to accommodate increased metabolic demands, a phenomenon known as functional sympatholysis. In inherited myopathies such as the dystrophinopathies Duchenne and Becker muscle dystrophies (DMD and BMD), nNOS is lost from the sarcolemma. The loss of nNOS may cause functional ischemia contributing to skeletal and cardiac muscle cell injury. Effects of NO is augmented by inhibiting degradation of the second messenger cyclic guanosine monophosphate (cGMP) using sildenafil and tadalafil, both of which inhibit the enzyme phosphodiesterase 5 (PDE5). In animal models of DMD, PDE5-inhibitors prevent functional ischemia, reduce post-exercise skeletal muscle pathology and fatigue, show amelioration of cardiac muscle cell damage and increase cardiac performance. However, effect on clinical outcomes in DMD and BMD patients have been disappointing with minor effects on upper limb performance and none on ambulation. This review aims to summarize the current knowledge of nNOS function related to functional sympatholysis in skeletal muscle and studies on PDE5-inhibitor treatment in nNOS-deficient animal models and patients.
Collapse
Affiliation(s)
- Nanna W Dombernowsky
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Joakim N E Ölmestig
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark
| | - Nanna Witting
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Christina Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark; PDE Research Group, Lundbeck Foundation Center for Neurovascular Research (LUCENS), Denmark.
| |
Collapse
|
13
|
Heydemann A. Skeletal Muscle Metabolism in Duchenne and Becker Muscular Dystrophy-Implications for Therapies. Nutrients 2018; 10:nu10060796. [PMID: 29925809 PMCID: PMC6024668 DOI: 10.3390/nu10060796] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 02/06/2023] Open
Abstract
The interactions between nutrition and metabolism and skeletal muscle have long been known. Muscle is the major metabolic organ—it consumes more calories than other organs—and therefore, there is a clear need to discuss these interactions and provide some direction for future research areas regarding muscle pathologies. In addition, new experiments and manuscripts continually reveal additional highly intricate, reciprocal interactions between metabolism and muscle. These reciprocal interactions include exercise, age, sex, diet, and pathologies including atrophy, hypoxia, obesity, diabetes, and muscle myopathies. Central to this review are the metabolic changes that occur in the skeletal muscle cells of muscular dystrophy patients and mouse models. Many of these metabolic changes are pathogenic (inappropriate body mass changes, mitochondrial dysfunction, reduced adenosine triphosphate (ATP) levels, and increased Ca2+) and others are compensatory (increased phosphorylated AMP activated protein kinase (pAMPK), increased slow fiber numbers, and increased utrophin). Therefore, reversing or enhancing these changes with therapies will aid the patients. The multiple therapeutic targets to reverse or enhance the metabolic pathways will be discussed. Among the therapeutic targets are increasing pAMPK, utrophin, mitochondrial number and slow fiber characteristics, and inhibiting reactive oxygen species. Because new data reveals many additional intricate levels of interactions, new questions are rapidly arising. How does muscular dystrophy alter metabolism, and are the changes compensatory or pathogenic? How does metabolism affect muscular dystrophy? Of course, the most profound question is whether clinicians can therapeutically target nutrition and metabolism for muscular dystrophy patient benefit? Obtaining the answers to these questions will greatly aid patients with muscular dystrophy.
Collapse
Affiliation(s)
- Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Center for Cardiovascular Research, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
14
|
Relation of nNOS isoforms to mitochondrial density and PGC-1alpha expression in striated muscles of mice. Nitric Oxide 2018; 77:35-43. [PMID: 29678764 DOI: 10.1016/j.niox.2018.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/10/2018] [Accepted: 04/16/2018] [Indexed: 01/16/2023]
Abstract
The expression of neuronal NO synthase (nNOS) alpha- and beta-isoforms in skeletal muscle is well documented but only little information is available about their regulation/functions. Using different mouse models, we now assessed whether the expression of nNOS-isoforms in muscle fibers is related to mitochondria content/activity and regulated by peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha). Catalytic histochemistry revealed highest nNOS-concentrations to be present in type-2 oxidative muscle fibers. Differences in mitochondrial density between nNOS-KO-mice and WT-littermates established by morphometry after transmission electron microscopy were significant in the oxidative portion of the tibialis anterior muscle (TA) but not in rectus femoris muscle (RF) indicating an nNOS-dependent mitochondrial pool in TA. Quantitative immunoblotting displayed the nNOS alpha-isoform to preponderate in those striated muscles of C57BL/6-mice that comprise of many type-2 oxidative fibers, e.g. TA, while roughly even levels of the two nNOS-isoforms were expressed in those muscles that mainly consist of type-2 glycolytic fibers, e.g. RF. Differences in citrate synthase-activity in muscle homogenates between nNOS-KO-mice and WT-littermates were positively related to nNOS alpha-isoform levels. In transgenic-mice over-expressing muscular PGC-1alpha compared to WT-littermates, immunoblotting revealed a significant shift in nNOS-expression in favor of the alpha-isoform in six out of eight striated muscles (exceptions: soleus muscle and tongue) without consistent relationship to changes in the expression of mitochondrial markers. In summary, our study demonstrated the nNOS alpha-isoform expression to be related to mitochondrial content/activity and to be up-regulated by up-stream PGC-1alpha in striated muscles, particularly in those enriched with type-2 oxidative fibers implying a functional convergence of the two signaling systems in these fibers.
Collapse
|
15
|
Wehling-Henricks M, Welc SS, Samengo G, Rinaldi C, Lindsey C, Wang Y, Lee J, Kuro-O M, Tidball JG. Macrophages escape Klotho gene silencing in the mdx mouse model of Duchenne muscular dystrophy and promote muscle growth and increase satellite cell numbers through a Klotho-mediated pathway. Hum Mol Genet 2018; 27:14-29. [PMID: 29040534 PMCID: PMC5886268 DOI: 10.1093/hmg/ddx380] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/29/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscle wasting disease in which inflammation influences the severity of pathology. We found that the onset of muscle inflammation in the mdx mouse model of DMD coincides with large increases in expression of pro-inflammatory cytokines [tumor necrosis factor-α (TNFα); interferon gamma (IFNγ)] and dramatic reductions of the pro-myogenic protein Klotho in muscle cells and large increases of Klotho in pro-regenerative, CD206+ macrophages. Furthermore, TNFα and IFNγ treatments reduced Klotho in muscle cells and increased Klotho in macrophages. Because CD206+/Klotho+ macrophages were concentrated at sites of muscle regeneration, we tested whether macrophage-derived Klotho promotes myogenesis. Klotho transgenic macrophages had a pro-proliferative influence on muscle cells that was ablated by neutralizing antibodies to Klotho and conditioned media from Klotho mutant macrophages did not increase muscle cell proliferation in vitro. In addition, transplantation of bone marrow cells from Klotho transgenic mice into mdx recipients increased numbers of myogenic cells and increased the size of muscle fibers. Klotho also acted directly on macrophages, stimulating their secretion of TNFα. Because TNFα is a muscle mitogen, we tested whether the pro-proliferative effects of Klotho on muscle cells were mediated by TNFα and found that increased proliferation caused by Klotho was reduced by anti-TNFα. Collectively, these data show that pro-inflammatory cytokines contribute to silencing of Klotho in dystrophic muscle, but increase Klotho expression by macrophages. Our findings also show that macrophage-derived Klotho can promote muscle regeneration by expanding populations of muscle stem cells and increasing muscle fiber growth in dystrophic muscle.
Collapse
Affiliation(s)
- Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Steven S Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Guiseppina Samengo
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Chiara Rinaldi
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Catherine Lindsey
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Ying Wang
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095, USA
| | - Jeongyoon Lee
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Yakushiji, Shimotsuke, Tochigi, Japan
| | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Costas-Insua C, Merino-Gracia J, Aicart-Ramos C, Rodríguez-Crespo I. Subcellular Targeting of Nitric Oxide Synthases Mediated by Their N-Terminal Motifs. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 111:165-195. [PMID: 29459031 DOI: 10.1016/bs.apcsb.2017.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
From a catalytic point of view, the three mammalian nitric oxide synthases (NOSs) function in an almost identical way. The N-terminal oxygenase domain catalyzes the conversion of l-arginine to l-citrulline plus ·NO in two sequential oxidation steps. Once l-arginine binds to the active site positioned above the heme moiety, two consecutive monooxygenation reactions take place. In the first step, l-arginine is hydroxylated to make Nω-hydroxy-l-arginine in a process that requires 1 molecule of NADPH and 1 molecule of O2 per mol of l-arginine reacted. In the second step, Nω-hydroxy-l-arginine, never leaving the active site, is oxidized to ·NO plus l-citrulline and 1 molecule of O2 and 0.5 molecules of NADPH are consumed. Since nitric oxide is an important signaling molecule that participates in a number of biological processes, including neurotransmission, vasodilation, and immune response, synthesis and release of ·NO in vivo must be exquisitely regulated both in time and in space. Hence, NOSs have evolved introducing in their amino acid sequences subcellular targeting motifs, most of them located at their N-termini. Deletion studies performed on recombinant, purified NOSs have revealed that part of the N-terminus of all three NOS can be eliminated with the resulting mutant enzymes still being catalytically active. Likewise, NOS isoforms lacking part of their N-terminus when transfected in cells render mislocalized, active proteins. In this review we will comment on the current knowledge of these subcellular targeting signals present in nNOS, iNOS, and eNOS.
Collapse
|
17
|
Timpani CA, Hayes A, Rybalka E. Therapeutic strategies to address neuronal nitric oxide synthase deficiency and the loss of nitric oxide bioavailability in Duchenne Muscular Dystrophy. Orphanet J Rare Dis 2017; 12:100. [PMID: 28545481 PMCID: PMC5445371 DOI: 10.1186/s13023-017-0652-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 05/12/2017] [Indexed: 12/25/2022] Open
Abstract
Duchenne Muscular Dystrophy is a rare and fatal neuromuscular disease in which the absence of dystrophin from the muscle membrane induces a secondary loss of neuronal nitric oxide synthase and the muscles capacity for endogenous nitric oxide synthesis. Since nitric oxide is a potent regulator of skeletal muscle metabolism, mass, function and regeneration, the loss of nitric oxide bioavailability is likely a key contributor to the chronic pathological wasting evident in Duchenne Muscular Dystrophy. As such, various therapeutic interventions to re-establish either the neuronal nitric oxide synthase protein deficit or the consequential loss of nitric oxide synthesis and bioavailability have been investigated in both animal models of Duchenne Muscular Dystrophy and in human clinical trials. Notably, the efficacy of these interventions are varied and not always translatable from animal model to human patients, highlighting a complex interplay of factors which determine the downstream modulatory effects of nitric oxide. We review these studies herein.
Collapse
Affiliation(s)
- Cara A Timpani
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001.,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia
| | - Alan Hayes
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001.,Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia
| | - Emma Rybalka
- College of Health & Biomedicine, Victoria University, PO Box 14428, Melbourne, Victoria, Australia, 8001. .,Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia. .,Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, 3021, Australia.
| |
Collapse
|
18
|
Timpani CA, Trewin AJ, Stojanovska V, Robinson A, Goodman CA, Nurgali K, Betik AC, Stepto N, Hayes A, McConell GK, Rybalka E. Attempting to Compensate for Reduced Neuronal Nitric Oxide Synthase Protein with Nitrate Supplementation Cannot Overcome Metabolic Dysfunction but Rather Has Detrimental Effects in Dystrophin-Deficient mdx Muscle. Neurotherapeutics 2017; 14:429-446. [PMID: 27921261 PMCID: PMC5398978 DOI: 10.1007/s13311-016-0494-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Duchenne muscular dystrophy arises from the loss of dystrophin and is characterized by calcium dysregulation, muscular atrophy, and metabolic dysfunction. The secondary reduction of neuronal nitric oxide synthase (nNOS) from the sarcolemma reduces NO production and bioavailability. As NO modulates glucose uptake, metabolism, and mitochondrial bioenergetics, we investigated whether an 8-week nitrate supplementation regimen could overcome metabolic dysfunction in the mdx mouse. Dystrophin-positive control (C57BL/10) and dystrophin-deficient mdx mice were supplemented with sodium nitrate (85 mg/l) in drinking water. Following the supplementation period, extensor digitorum longus and soleus were excised and radioactive glucose uptake was measured at rest (basal) and during contraction. Gastrocnemius was excised and mitochondrial respiration was measured using the Oroboros Oxygraph. Tibialis anterior was analyzed immunohistochemically for the presence of dystrophin, nNOS, nitrotyrosine, IgG and CD45+ cells, and histologically to assess areas of damage and regeneration. Glucose uptake in the basal and contracting states was normal in unsupplemented mdx muscles but was reduced following nitrate supplementation in mdx muscles only. The mitochondrial utilization of substrates was also impaired in mdx gastrocnemius during phosphorylating and maximal uncoupled respiration, and nitrate could not improve respiration in mdx muscle. Although nitrate supplementation reduced mitochondrial hydrogen peroxide emission, it induced mitochondrial uncoupling in red gastrocnemius, increased muscle fiber peroxynitrite (nitrotyrosine), and promoted skeletal muscle damage. Our novel data suggest that despite lower nNOS protein expression and likely lower NO production in mdx muscle, enhancing NO production with nitrate supplementation in these mice has detrimental effects on skeletal muscle. This may have important relevance for those with DMD.
Collapse
Affiliation(s)
- Cara A Timpani
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Adam J Trewin
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
| | - Vanesa Stojanovska
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Ainsley Robinson
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Craig A Goodman
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Health, Melbourne, Victoria, 3021, Australia
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Andrew C Betik
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
| | - Nigel Stepto
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
| | - Alan Hayes
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Health, Melbourne, Victoria, 3021, Australia
| | - Glenn K McConell
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Health, Melbourne, Victoria, 3021, Australia
| | - Emma Rybalka
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia.
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia.
- Australian Institute of Musculoskeletal Science (AIMSS), Western Health, Melbourne, Victoria, 3021, Australia.
| |
Collapse
|
19
|
Zhang F, Qian X, Qin C, Lin Y, Wu H, Chang L, Luo C, Zhu D. Phosphofructokinase-1 Negatively Regulates Neurogenesis from Neural Stem Cells. Neurosci Bull 2016; 32:205-16. [PMID: 27146165 DOI: 10.1007/s12264-016-0032-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/13/2016] [Indexed: 12/31/2022] Open
Abstract
Phosphofructokinase-1 (PFK-1), a major regulatory glycolytic enzyme, has been implicated in the functions of astrocytes and neurons. Here, we report that PFK-1 negatively regulates neurogenesis from neural stem cells (NSCs) by targeting pro-neural transcriptional factors. Using in vitro assays, we found that PFK-1 knockdown enhanced, and PFK-1 overexpression inhibited the neuronal differentiation of NSCs, which was consistent with the findings from NSCs subjected to 5 h of hypoxia. Meanwhile, the neurogenesis induced by PFK-1 knockdown was attributed to the increased proliferation of neural progenitors and the commitment of NSCs to the neuronal lineage. Similarly, in vivo knockdown of PFK-1 also increased neurogenesis in the dentate gyrus of the hippocampus. Finally, we demonstrated that the neurogenesis mediated by PFK-1 was likely achieved by targeting mammalian achaete-scute homologue-1 (Mash 1), neuronal differentiation factor (NeuroD), and sex-determining region Y (SRY)-related HMG box 2 (Sox2). All together, our results reveal PFK-1 as an important regulator of neurogenesis.
Collapse
Affiliation(s)
- Fengyun Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaodan Qian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Qin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yuhui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Haiyin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Chunxia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.,Institute of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, China.,Laboratory of Cerebrovascular Disease, Nanjing Medical University, Nanjing, 211166, China
| | - Dongya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China. .,Institute of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, China. .,Laboratory of Cerebrovascular Disease, Nanjing Medical University, Nanjing, 211166, China. .,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, 211166, China.
| |
Collapse
|
20
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Massimino ML, Peggion C, Loro F, Stella R, Megighian A, Scorzeto M, Blaauw B, Toniolo L, Sorgato MC, Reggiani C, Bertoli A. Age-dependent neuromuscular impairment in prion protein knockout mice. Muscle Nerve 2015; 53:269-79. [DOI: 10.1002/mus.24708] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2015] [Indexed: 12/26/2022]
Affiliation(s)
| | - Caterina Peggion
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Federica Loro
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Roberto Stella
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Aram Megighian
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Michele Scorzeto
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Bert Blaauw
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Luana Toniolo
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Maria Catia Sorgato
- CNR Neuroscience Institute, University of Padova
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Carlo Reggiani
- CNR Neuroscience Institute, University of Padova
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| | - Alessandro Bertoli
- Department of Biomedical Sciences; University of Padova; Via U. Bassi 58/B 35131 Padova Italy
| |
Collapse
|
22
|
Liu DZ, Stamova B, Hu S, Ander BP, Jickling GC, Zhan X, Sharp FR, Wong B. MicroRNA and mRNA Expression Changes in Steroid Naïve and Steroid Treated DMD Patients. J Neuromuscul Dis 2015; 2:387-396. [PMID: 27858746 PMCID: PMC5240570 DOI: 10.3233/jnd-150076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background: Duchenne Muscular Dystrophy (DMD) is a recessive X-linked form of muscular dystrophy. Steroid therapy has clinical benefits for DMD patients, but the mechanism remains unclear. Objective: This study was designed to identify mRNAs and microRNAs regulated in Duchenne Muscular Dystrophy patients prior to and after steroid therapy. Methods: Genome wide transcriptome profiling of whole blood was performed to identify mRNAs and microRNAs regulated in DMD patients. Results: The data show many regulated mRNAs and some microRNAs, including some muscle-specific microRNAs (e.g., miR-206), that were significantly altered in blood of young (age 3–10) DMD patients compared to young controls. A total of 95 microRNAs, but no mRNAs, were differentially expressed in older DMD patients compared to matched controls (age 11–20). Steroid treatment reversed expression patterns of several microRNAs (miR-206, miR-181a, miR-4538, miR-4539, miR-606, and miR-454) that were altered in the young DMD patients. As an example, the over-expression of miR-206 in young DMD patients is predicted to down-regulate a set of target genes (e.g., RHGAP31, KHSRP, CORO1B, PTBP1, C7orf58, DLG4, and KLF4) that would worsen motor function. Since steroids decreased miR-206 expression to control levels, this could provide one mechanism by which steroids improve motor function. Conclusions: These identified microRNA-mRNA alterations will help better understand the pathophysiology of DMD and the response to steroid treatment.
Collapse
Affiliation(s)
- Da Zhi Liu
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Boryana Stamova
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Shengyong Hu
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bradley P Ander
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Glen C Jickling
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Xinhua Zhan
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Frank R Sharp
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, California, USA
| | - Brenda Wong
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
23
|
Revisiting the dystrophin-ATP connection: How half a century of research still implicates mitochondrial dysfunction in Duchenne Muscular Dystrophy aetiology. Med Hypotheses 2015; 85:1021-33. [PMID: 26365249 DOI: 10.1016/j.mehy.2015.08.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/24/2015] [Indexed: 12/22/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal neuromuscular disease that is characterised by dystrophin-deficiency and chronic Ca(2+)-induced skeletal muscle wasting, which currently has no cure. DMD was once considered predominantly as a metabolic disease due to the myriad of metabolic insufficiencies evident in the musculature, however this aspect of the disease has been extensively ignored since the discovery of dystrophin. The collective historical and contemporary literature documenting these metabolic nuances has culminated in a series of studies that importantly demonstrate that metabolic dysfunction exists independent of dystrophin expression and a mild disease phenotype can be expressed even in the complete absence of dystrophin expression. Targeting and supporting metabolic pathways with anaplerotic and other energy-enhancing supplements has also shown therapeutic value. We explore the hypothesis that DMD is characterised by a systemic mitochondrial impairment that is central to disease aetiology rather than a secondary pathophysiological consequence of dystrophin-deficiency.
Collapse
|
24
|
Messina S, Bitto A, Vita GL, Aguennouz M, Irrera N, Licata N, Sframeli M, Bruschetta D, Minutoli L, Altavilla D, Vita G, Squadrito F. Modulation of neuronal nitric oxide synthase and apoptosis by the isoflavone genistein in Mdx mice. Biofactors 2015; 41:324-9. [PMID: 26332024 DOI: 10.1002/biof.1226] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/13/2015] [Accepted: 07/23/2015] [Indexed: 11/08/2022]
Abstract
Dystrophin lack in DMD causes neuronal nitric oxide synthase (nNOS) membrane delocalization which in turn promotes functional muscle ischemia, and exacerbates muscle injury. Apoptosis and the exhaustion of muscle regenerative capacity are implicated in Duchenne muscular dystrophy (DMD) pathogenesis and therefore are relevant therapeutic targets. Genistein has been reported to have pro-proliferative effects, promoting G1/S cell phase transition through the induction of cyclin D1, and anti-apoptotic properties. We previously showed that genistein could reduce muscle necrosis and enhance regeneration with an augmented number of myogenin-positive satellite cells and myonuclei, ameliorating muscle function in mdx mice. In this study we evaluated the underlying mechanisms of genistein effect on muscle specimens of mdx and wild type mice treated for five weeks with genistein (2 mg/kg/i.p. daily) or vehicle. Western blot analysis show that genistein increased cyclin D1 and nNOS expression; and showed an antiapoptotic effect, modulating the expression of BAX and Bcl-2. Our results suggest that this isoflavone might enhance the regenerative spurt in mdx mice muscle restoring nNOS, promoting G1/S phase transition in muscle cell, and inhibiting apoptosis. Further studies with longer time treatment or using different experimental approaches are needed to better investigate the underlying mechanisms of such results.
Collapse
Affiliation(s)
- Sonia Messina
- Department of Neurosciences, University of Messina, Messina, Italy
- Centro Clinico Nemo Sud, Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, Messina, Italy
| | - Gian Luca Vita
- Department of Neurosciences, University of Messina, Messina, Italy
- Centro Clinico Nemo Sud, Messina, Italy
| | | | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, Messina, Italy
| | - Norma Licata
- Department of Neurosciences, University of Messina, Messina, Italy
| | | | - Daniele Bruschetta
- Department of Biomorphology and Biotechnologies, University of Messina, Messina, Italy
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, Messina, Italy
| | - Domenica Altavilla
- Department of Paediatric, Gynaecological, Microbiological and Biomedical Sciences, University of Messina, Messina, Italy
| | - Giuseppe Vita
- Department of Neurosciences, University of Messina, Messina, Italy
- Centro Clinico Nemo Sud, Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, Messina, Italy
| |
Collapse
|
25
|
Naproxcinod shows significant advantages over naproxen in the mdx model of Duchenne Muscular Dystrophy. Orphanet J Rare Dis 2015; 10:101. [PMID: 26296873 PMCID: PMC4546261 DOI: 10.1186/s13023-015-0311-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/23/2015] [Indexed: 12/18/2022] Open
Abstract
Background In dystrophin-deficient muscles of Duchenne Muscular Dystrophy (DMD) patients and the mdx mouse model, nitric oxide (NO) signalling is impaired. Previous studies have shown that NO-donating drugs are beneficial in dystrophic mouse models. Recently, a long-term treatment (9 months) of mdx mice with naproxcinod, an NO-donating naproxen, has shown a significant improvement of the dystrophic phenotype with beneficial effects present throughout the disease progression. It remains however to be clearly dissected out which specific effects are due to the NO component compared with the anti-inflammatory activity associated with naproxen. Understanding the contribution of NO vs the anti-inflammatory effect is important, in view of the potential therapeutic perspective, and this is the final aim of this study. Methods Five-week-old mdx mice received either naproxcinod (30 mg/kg) or the equimolar dose of naproxen (20 mg/kg) in the diet for 6 months. Control mdx mice were used as reference. Treatments (or vehicle for control groups) were administered daily in the diet. For the first 3 months the study was performed in sedentary animals, then all mice were subjected to exercise until the sixth month. Skeletal muscle force was assessed by measuring whole body tension in sedentary animals as well as in exercised mice and resistance to fatigue was measured after 3 months of running exercise. At the end of 6 months of treatment, animals were sacrificed for histological analysis and measurement of naproxen levels in blood and skeletal muscle. Results Naproxcinod significantly ameliorated skeletal muscle force and resistance to fatigue in sedentary as well as in exercised mice, reduced inflammatory infiltrates and fibrosis deposition in both cardiac and diaphragm muscles. Conversely, the equimolar dose of naproxen showed no effects on fibrosis and improved muscle function only in sedentary mice, while the beneficial effects in exercised mice were lost demonstrating a limited and short-term effect. Conclusion In conclusion, this study shows that NO donation may have an important role, in addition to anti-inflammatory activity, in slowing down the progression of the disease in the mdx mouse model therefore positioning naproxcinod as a promising candidate for treatment of DMD. Electronic supplementary material The online version of this article (doi:10.1186/s13023-015-0311-0) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Dystrophin induced cognitive impairment: mechanisms, models and therapeutic strategies. Ann Neurosci 2015; 22:108-18. [PMID: 26130916 PMCID: PMC4480258 DOI: 10.5214/ans.0972.7531.221210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/19/2015] [Accepted: 03/05/2015] [Indexed: 12/13/2022] Open
Abstract
Existence of conserved domains in dystrophin and its associated complexes provide an opportunity to understand the role of dystrophin associated signalling and its association with neuronal metabolism in a variety of model organisms. We critically reviewed the studies till 2013 through established search engines and databases. Thus, we review the role of dystrophin and its isoforms in different animal models at developmental stages in the neuronal metabolism to enhance the therapeutic strategies. Dystrophin interacts with other proteins in such a way that, when affected, it results in co-morbidities including autism and other neuropsychiatric disorders. It is speculated that various signalling molecules may converge to disrupt neuronal metabolism not adequately studied. TGF-β, RhoGAP and CAM mediated signalling molecules are the chief cause of mortalities due to respiratory and cardiac involvement but remain underevaluated targets for cognitive impairment in DMD/BMD. Manipulation of these signalling pathways could be potent intervention in dystrophin induced cognitive impairment while complementary therapeutic approaches may also be helpful in the treatment of cognitive impairment associated with DMD/BMD.
Collapse
|
27
|
De Palma C, Morisi F, Pambianco S, Assi E, Touvier T, Russo S, Perrotta C, Romanello V, Carnio S, Cappello V, Pellegrino P, Moscheni C, Bassi MT, Sandri M, Cervia D, Clementi E. Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation. Skelet Muscle 2014; 4:22. [PMID: 25530838 PMCID: PMC4272808 DOI: 10.1186/s13395-014-0022-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/18/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Nitric oxide (NO), generated in skeletal muscle mostly by the neuronal NO synthases (nNOSμ), has profound effects on both mitochondrial bioenergetics and muscle development and function. The importance of NO for muscle repair emerges from the observation that nNOS signalling is defective in many genetically diverse skeletal muscle diseases in which muscle repair is dysregulated. How the effects of NO/nNOSμ on mitochondria impact on muscle function, however, has not been investigated yet. METHODS In this study we have examined the relationship between the NO system, mitochondrial structure/activity and skeletal muscle phenotype/growth/functions using a mouse model in which nNOSμ is absent. Also, NO-induced effects and the NO pathway were dissected in myogenic precursor cells. RESULTS We show that nNOSμ deficiency in mouse skeletal muscle leads to altered mitochondrial bioenergetics and network remodelling, and increased mitochondrial unfolded protein response (UPR(mt)) and autophagy. The absence of nNOSμ is also accompanied by an altered mitochondrial homeostasis in myogenic precursor cells with a decrease in the number of myonuclei per fibre and impaired muscle development at early stages of perinatal growth. No alterations were observed, however, in the overall resting muscle structure, apart from a reduced specific muscle mass and cross sectional areas of the myofibres. Investigating the molecular mechanisms we found that nNOSμ deficiency was associated with an inhibition of the Akt-mammalian target of rapamycin pathway. Concomitantly, the Akt-FoxO3-mitochondrial E3 ubiquitin protein ligase 1 (Mul-1) axis was also dysregulated. In particular, inhibition of nNOS/NO/cyclic guanosine monophosphate (cGMP)/cGMP-dependent-protein kinases induced the transcriptional activity of FoxO3 and increased Mul-1 expression. nNOSμ deficiency was also accompanied by functional changes in muscle with reduced muscle force, decreased resistance to fatigue and increased degeneration/damage post-exercise. CONCLUSIONS Our results indicate that nNOSμ/NO is required to regulate key homeostatic mechanisms in skeletal muscle, namely mitochondrial bioenergetics and network remodelling, UPR(mt) and autophagy. These events are likely associated with nNOSμ-dependent impairments of muscle fibre growth resulting in a deficit of muscle performance.
Collapse
Affiliation(s)
- Clara De Palma
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Federica Morisi
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Sarah Pambianco
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Emma Assi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Thierry Touvier
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Stefania Russo
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Cristiana Perrotta
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Vanina Romanello
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
| | - Silvia Carnio
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
| | - Valentina Cappello
- National Research Council-Institute of Neuroscience, Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milano, Italy ; CNI@NEST, Italian Institute of Technology, Pisa, Italy
| | - Paolo Pellegrino
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Claudia Moscheni
- Unit of Morphology, Department of Biomedical and Clinical Sciences "Luigi Sacco", Università di Milano, Milano, Italy
| | | | - Marco Sandri
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy ; Department of Biomedical Science, Università di Padova, Padova, Italy
| | - Davide Cervia
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy ; Department for Innovation in Biological, Agro-food and Forest Systems, Università della Tuscia, Viterbo, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy ; Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| |
Collapse
|
28
|
Froehner SC, Reed SM, Anderson KN, Huang PL, Percival JM. Loss of nNOS inhibits compensatory muscle hypertrophy and exacerbates inflammation and eccentric contraction-induced damage in mdx mice. Hum Mol Genet 2014; 24:492-505. [PMID: 25214536 DOI: 10.1093/hmg/ddu469] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Approaches targeting nitric oxide (NO) signaling show promise as therapies for Duchenne and Becker muscular dystrophies. However, the mechanisms by which NO benefits dystrophin-deficient muscle remain unclear, but may involve nNOSβ, a newly discovered enzymatic source of NO in skeletal muscle. Here we investigate the impact of dystrophin deficiency on nNOSβ and use mdx mice engineered to lack nNOSμ and nNOSβ to discern how the loss of nNOS impacts dystrophic skeletal muscle pathology. In mdx muscle, nNOSβ was mislocalized and its association with the Golgi complex was reduced. nNOS depletion from mdx mice prevented compensatory skeletal muscle cell hypertrophy, decreased myofiber central nucleation and increased focal macrophage cell infiltration, indicating exacerbated dystrophic muscle damage. Reductions in muscle integrity in nNOS-null mdx mice were accompanied by decreases in specific force and increased susceptibility to eccentric contraction-induced muscle damage compared with mdx controls. Unexpectedly, muscle fatigue was unaffected by nNOS depletion, revealing a novel latent compensatory mechanism for the loss of nNOS in mdx mice. Together with previous studies, these data suggest that localization of both nNOSμ and nNOSβ is disrupted by dystrophin deficiency. They also indicate that nNOS has a more complex role as a modifier of dystrophic pathology and broader therapeutic potential than previously recognized. Importantly, these findings also suggest nNOSβ as a new drug target and provide a new conceptual framework for understanding nNOS signaling and the benefits of NO therapies in dystrophinopathies.
Collapse
Affiliation(s)
- Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington Medical School, Seattle, WA, USA
| | - Sarah M Reed
- Department of Physiology and Biophysics, University of Washington Medical School, Seattle, WA, USA
| | - Kendra N Anderson
- Department of Physiology and Biophysics, University of Washington Medical School, Seattle, WA, USA
| | - Paul L Huang
- Cardiovascular Research Center and Harvard Stem Cell Institute, Massachusetts General Hospital, Boston, MA, USA and
| | - Justin M Percival
- Department of Physiology and Biophysics, University of Washington Medical School, Seattle, WA, USA Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
29
|
Tidball JG, Wehling-Henricks M. Nitric oxide synthase deficiency and the pathophysiology of muscular dystrophy. J Physiol 2014; 592:4627-38. [PMID: 25194047 DOI: 10.1113/jphysiol.2014.274878] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The secondary loss of neuronal nitric oxide synthase (nNOS) that occurs in dystrophic muscle is the basis of numerous, complex and interacting features of the dystrophic pathology that affect not only muscle itself, but also influence the interaction of muscle with other tissues. Many mechanisms through which nNOS deficiency contributes to misregulation of muscle development, blood flow, fatigue, inflammation and fibrosis in dystrophic muscle have been identified, suggesting that normalization in NO production could greatly attenuate diverse aspects of the pathology of muscular dystrophy through multiple regulatory pathways. However, the relative importance of the loss of nNOS from the sarcolemma versus the importance of loss of total nNOS from dystrophic muscle remains unknown. Although most current evidence indicates that nNOS localization at the sarcolemma is not required to achieve NO-mediated reductions of pathology in muscular dystrophy, the question remains open concerning whether membrane localization would provide a more efficient rescue from features of the dystrophic phenotype.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, USA Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
30
|
Cossu MV, Cattaneo D, Fucile S, Pellegrino P, Baldelli S, Cozzi V, Capetti A, Clementi E. Combined isosorbide dinitrate and ibuprofen as a novel therapy for muscular dystrophies: evidence from Phase I studies in healthy volunteers. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:411-9. [PMID: 24851040 PMCID: PMC4018313 DOI: 10.2147/dddt.s58803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We designed two Phase I studies that assessed healthy volunteers in order to evaluate the safety and to optimize the dosing of the combination of the drugs isosorbide dinitrate, a nitric oxide donor, and ibuprofen, a nonsteroidal antiinflammatory drug. We designed these studies with the aim of designing a Phase II trial to evaluate the drugs’ efficacy in patients affected by Duchenne muscular dystrophy. For the first trial, ISOFEN1, a single-dose, randomized-sequence, open-label, active control, three-treatment cross-over study, was aimed at comparing the pharmacokinetics of ibuprofen 200 mg and isosorbide dinitrate 20 mg when given alone and concomitantly. The pharmacokinetics of ibuprofen given alone versus ibuprofen given concomitantly with isosorbide dinitrate were similar, as documented by the lack of statistically significant differences in the main drug’s pharmacokinetic parameters (time to maximal concentration [Tmax], maximal concentration [Cmax], area under the curve [AUC]0–t, and AUC0–∞). Similarly, we found that the coadministration of ibuprofen did not significantly affect the pharmacokinetics of isosorbide dinitrate. No issues of safety were detected. The second trial, ISOFEN2, was a single-site, dose titration study that was designed to select the maximum tolerated dose for isosorbide dinitrate when coadministered with ibuprofen. Eighteen out of the 19 enrolled subjects tolerated the treatment well, and they completed the study at the highest dose of isosorbide dinitrate applied (80 mg/day). One subject voluntarily decided to reduce the dose of isosorbide dinitrate from 80 mg to 60 mg. The treatment-related adverse events recorded during the study were, for the large majority, episodes of headache that remitted spontaneously in 0.5–1 hour – a known side effect of isosorbide dinitrate. These studies demonstrate that the combination of isosorbide dinitrate and ibuprofen does not lead to pharmacokinetic interactions between the two drugs; they also demonstrate that the combination of isosorbide dinitrate and ibuprofen has optimal tolerability and safety profiles that are similar to those previously reported for isosorbide dinitrate and ibuprofen given alone.
Collapse
Affiliation(s)
- Maria Vittoria Cossu
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Serena Fucile
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Paolo Pellegrino
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Valeria Cozzi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Amedeo Capetti
- Unit of Infectious Diseases, University Hospital "Luigi Sacco", Milan, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy ; Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy
| |
Collapse
|
31
|
Panda SP, Li W, Venkatakrishnan P, Chen L, Astashkin AV, Masters BSS, Feng C, Roman LJ. Differential calmodulin-modulatory and electron transfer properties of neuronal nitric oxide synthase mu compared to the alpha variant. FEBS Lett 2013; 587:3973-8. [PMID: 24211446 DOI: 10.1016/j.febslet.2013.10.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 10/26/2022]
Abstract
Neuronal nitric oxide synthase μ (nNOSμ) contains 34 additional residues in an autoregulatory element compared to nNOSα. Cytochrome c and flavin reductions in the absence of calmodulin (CaM) were faster in nNOSμ than nNOSα, while rates in the presence of CaM were smaller. The magnitude of stimulation by CaM is thus notably lower in nNOSμ. No difference in NO production was observed, while electron transfer between the FMN and heme moieties and formation of an inhibitory ferrous-nitrosyl complex were slower in nNOSμ. Thus, the insert affects electron transfer rates, modulation of electron flow by CaM, and heme-nitrosyl complex formation.
Collapse
Affiliation(s)
- Satya P Panda
- Department of Biochemistry, University of Texas Health Science Center in San Antonio, San Antonio, TX 78229, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wary C, Naulet T, Thibaud JL, Monnet A, Blot S, Carlier PG. Splitting of Pi and other ³¹P NMR anomalies of skeletal muscle metabolites in canine muscular dystrophy. NMR IN BIOMEDICINE 2012; 25:1160-1169. [PMID: 22354667 DOI: 10.1002/nbm.2785] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 12/16/2011] [Accepted: 01/10/2012] [Indexed: 05/27/2023]
Abstract
Many anomalies exist in the resting (31) P muscle spectra of boys with Duchenne muscular dystrophy (DMD) but few have been reported in Golden Retriever muscular dystrophy (GRMD), the closest existing animal model for DMD. Because GRMD is recommended for preclinical evaluation of therapies and quantitative outcome measures are needed, we investigated anomalies of (31) P NMRS in tibial cranial and biceps femoris muscles from 14 GRMD compared to 9 control (CONT) dogs. Alterations observed in DMD children - low phosphocreatine and high phospho-monoesters and -diesters - were all found in GRMD but increased pH was not. More surprisingly, inorganic phosphate (Pi) appeared to present a prominent splitting with an enhanced Pi(b) resonance at 0.3 ppm downfield of Pi(a) . Assuming that both resonances are Pi, the pH for Pi(a) in GRMD corresponded to a physiological intracellular pH(a) (6.97 ± 0.05), while pH(b) approached the extracellular range (7.27 ± 0.10) and correlated with pH(a) in GRMD (R(2) = 0.65). Both Pi(a) and Pi(b) were elevated compared to CONT and Pi(a) increased with age for GRMD (R(2) = 0.48, p < 0.001). Magnetisation transfer experiments between γATP and Pi were conducted to better characterise Pi pools. Equal T1 relaxation times for Pi(b) and Pi(a) did not support a mitochondrial origin of Pi(b) . We suggest that Pi(b) could originate from degenerating hypercontracted cells that have a leaky membrane and inadequate cell homeostasis and pH regulation. Pi(b) showed minimal chemical exchange in all dogs, while the exchange rate of Pi(a) was reduced in GRMD and might extraneously reflect low glycolytic activity in DMD. Taken together, the ensemble of (31) P NMRS alterations identifies muscle dysfunction and could provide useful biomarkers of therapeutic efficacy. Furthermore, among these, two might relate more specifically to dystrophic processes and merit further investigation: one is the existence of the enhanced alkaline Pi(b) pool; the other, mechanisms by which membrane disruption might increase phosphodiesters in dystrophy.
Collapse
Affiliation(s)
- Claire Wary
- NMR Laboratory, Institute of Myology, Paris, France.
| | | | | | | | | | | |
Collapse
|
33
|
Percival JM, Whitehead NP, Adams ME, Adamo CM, Beavo JA, Froehner SC. Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy. J Pathol 2012; 228:77-87. [PMID: 22653783 DOI: 10.1002/path.4054] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/16/2010] [Accepted: 05/21/2010] [Indexed: 11/07/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy caused by mutations in the dystrophin gene. Loss of dystrophin initiates a progressive decline in skeletal muscle integrity and contractile capacity which weakens respiratory muscles including the diaphragm, culminating in respiratory failure, the leading cause of morbidity and mortality in DMD patients. At present, corticosteroid treatment is the primary pharmacological intervention in DMD, but has limited efficacy and adverse side effects. Thus, there is an urgent need for new safe, cost-effective, and rapidly implementable treatments that slow disease progression. One promising new approach is the amplification of nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signalling pathways with phosphodiesterase 5 (PDE5) inhibitors. PDE5 inhibitors serve to amplify NO signalling that is attenuated in many neuromuscular diseases including DMD. We report here that a 14-week treatment of the mdx mouse model of DMD with the PDE5 inhibitor sildenafil (Viagra(®), Revatio(®)) significantly reduced mdx diaphragm muscle weakness without impacting fatigue resistance. In addition to enhancing respiratory muscle contractility, sildenafil also promoted normal extracellular matrix organization. PDE5 inhibition slowed the establishment of mdx diaphragm fibrosis and reduced matrix metalloproteinase-13 (MMP-13) expression. Sildenafil also normalized the expression of the pro-fibrotic (and pro-inflammatory) cytokine tumour necrosis factor α (TNFα). Sildenafil-treated mdx diaphragms accumulated significantly less Evans Blue tracer dye than untreated controls, which is also indicative of improved diaphragm muscle health. We conclude that sildenafil-mediated PDE5 inhibition significantly reduces diaphragm respiratory muscle dysfunction and pathology in the mdx mouse model of Duchenne muscular dystrophy. This study provides new insights into the therapeutic utility of targeting defects in NO-cGMP signalling with PDE5 inhibitors in dystrophin-deficient muscle.
Collapse
Affiliation(s)
- Justin M Percival
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Buono R, Vantaggiato C, Pisa V, Azzoni E, Bassi MT, Brunelli S, Sciorati C, Clementi E. Nitric oxide sustains long-term skeletal muscle regeneration by regulating fate of satellite cells via signaling pathways requiring Vangl2 and cyclic GMP. Stem Cells 2012; 30:197-209. [PMID: 22084027 PMCID: PMC3378700 DOI: 10.1002/stem.783] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Satellite cells are myogenic precursors that proliferate, activate, and differentiate on muscle injury to sustain the regenerative capacity of adult skeletal muscle; in this process, they self-renew through the return to quiescence of the cycling progeny. This mechanism, while efficient in physiological conditions does not prevent exhaustion of satellite cells in pathologies such as muscular dystrophy where numerous rounds of damage occur. Here, we describe a key role of nitric oxide, an important signaling molecule in adult skeletal muscle, on satellite cells maintenance, studied ex vivo on isolated myofibers and in vivo using the α-sarcoglycan null mouse model of dystrophy and a cardiotoxin-induced model of repetitive damage. Nitric oxide stimulated satellite cells proliferation in a pathway dependent on cGMP generation. Furthermore, it increased the number of Pax7+/Myf5− cells in a cGMP-independent pathway requiring enhanced expression of Vangl2, a member of the planar cell polarity pathway involved in the Wnt noncanonical pathway. The enhanced self-renewal ability of satellite cells induced by nitric oxide is sufficient to delay the reduction of the satellite cell pool during repetitive acute and chronic damages, favoring muscle regeneration; in the α-sarcoglycan null dystrophic mouse, it also slowed disease progression persistently. These results identify nitric oxide as a key messenger in satellite cells maintenance, expand the significance of the Vangl2-dependent Wnt noncanonical pathway in myogenesis, and indicate novel strategies to optimize nitric oxide-based therapies for muscular dystrophy. Stem Cells 2012; 30:197–209.
Collapse
Affiliation(s)
- Roberta Buono
- Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
35
|
D'Angelo MG, Gandossini S, Martinelli Boneschi F, Sciorati C, Bonato S, Brighina E, Comi GP, Turconi AC, Magri F, Stefanoni G, Brunelli S, Bresolin N, Cattaneo D, Clementi E. Nitric oxide donor and non steroidal anti inflammatory drugs as a therapy for muscular dystrophies: evidence from a safety study with pilot efficacy measures in adult dystrophic patients. Pharmacol Res 2012; 65:472-9. [PMID: 22306844 DOI: 10.1016/j.phrs.2012.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/16/2012] [Accepted: 01/16/2012] [Indexed: 01/22/2023]
Abstract
This open-label, single centre pilot study was designed to evaluate safety and tolerability of the combination of the drugs isosorbide dinitrate, a nitric oxide donor, and ibuprofen, a non steroid anti-inflammatory drug, in a cohort of adult dystrophic patients (Duchenne, Becker and Limb-Girdle Muscular Dystrophy). Seventy-one patients were recruited: 35, treated with the drug combination for 12 months, and 36 untreated. Safety and adverse events were assessed by reported signs and symptoms, physical examinations, blood tests, cardiac and respiratory function tests. Exploratory outcomes measure, such as the motor function measure scale, were also applied. Good safety and tolerability profiles of the long-term co-administration of the drugs were demonstrated. Few and transient side effects (i.e. headache and low blood pressure) were reported. Additionally, exploratory outcomes measures were feasible in all the disease population studied and evidenced a trend towards amelioration that reached statistical significance in one dimension of the MFM scale. Systemic administration of ibuprofen and isosorbide dinitrate provides an adequate safety margin for clinical studies aimed at assessing efficacy.
Collapse
|
36
|
Schuh RA, Jackson KC, Khairallah RJ, Ward CW, Spangenburg EE. Measuring mitochondrial respiration in intact single muscle fibers. Am J Physiol Regul Integr Comp Physiol 2011; 302:R712-9. [PMID: 22160545 DOI: 10.1152/ajpregu.00229.2011] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Measurement of mitochondrial function in skeletal muscle is a vital tool for understanding regulation of cellular bioenergetics. Currently, a number of different experimental approaches are employed to quantify mitochondrial function, with each involving either mechanically or chemically induced disruption of cellular membranes. Here, we describe a novel approach that allows for the quantification of substrate-induced mitochondria-driven oxygen consumption in intact single skeletal muscle fibers isolated from adult mice. Specifically, we isolated intact muscle fibers from the flexor digitorum brevis muscle and placed the fibers in culture conditions overnight. We then quantified oxygen consumption rates using a highly sensitive microplate format. Peak oxygen consumption rates were significantly increased by 3.4-fold and 2.9-fold by simultaneous stimulation with the uncoupling agent, carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone (FCCP), and/or pyruvate or palmitate exposure, respectively. However, when calculating the total oxygen consumed over the entire treatment, palmitate exposure resulted in significantly more oxygen consumption compared with pyruvate. Further, as proof of principle for the procedure, we isolated fibers from the mdx mouse model, which has known mitochondrial deficits. We found significant reductions in initial and peak oxygen consumption of 51% and 61% compared with fibers isolated from the wild-type (WT) animals, respectively. In addition, we determined that fibers isolated from mdx mice exhibited less total oxygen consumption in response to the FCCP + pyruvate stimulation compared with the WT mice. This novel approach allows the user to make mitochondria-specific measures in a nondisrupted muscle fiber that has been isolated from a whole muscle.
Collapse
Affiliation(s)
- Rosemary A Schuh
- Research Service, Maryland Veterans Affairs Health Care System, Baltimore, Maryland 20742, USA
| | | | | | | | | |
Collapse
|
37
|
nNOS regulation of skeletal muscle fatigue and exercise performance. Biophys Rev 2011; 3:209-217. [PMID: 28510048 DOI: 10.1007/s12551-011-0060-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/17/2011] [Indexed: 10/15/2022] Open
Abstract
Neuronal nitric oxide synthases (nNOS) are Ca2+/calmodulin-activated enzymes that synthesize the gaseous messenger nitric oxide (NO). nNOSμ and the recently described nNOSβ, both spliced nNOS isoforms, are important enzymatic sources of NO in skeletal muscle, a tissue long considered to be a paradigmatic system for studying NO-dependent redox signaling. nNOS is indispensable for skeletal muscle integrity and contractile performance, and deregulation of nNOSμ signaling is a common pathogenic feature of many neuromuscular diseases. Recent evidence suggests that both nNOSμ and nNOSβ regulate skeletal muscle size, strength, and fatigue resistance, making them important players in exercise performance. nNOSμ acts as an activity sensor and appears to assist skeletal muscle adaptation to new functional demands, particularly those of endurance exercise. Prolonged inactivity leads to nNOS-mediated muscle atrophy through a FoxO-dependent pathway. nNOS also plays a role in modulating exercise performance in neuromuscular disease. In the mdx mouse model of Duchenne muscular dystrophy, defective nNOS signaling is thought to restrict contractile capacity of working muscle in two ways: loss of sarcolemmal nNOSμ causes excessive ischemic damage while residual cytosolic nNOSμ contributes to hypernitrosylation of the ryanodine receptor, causing pathogenic Ca2+ leak. This defect in Ca2+ handling promotes muscle damage, weakness, and fatigue. This review addresses these recent advances in the understanding of nNOS-dependent redox regulation of skeletal muscle function and exercise performance under physiological and neuromuscular disease conditions.
Collapse
|
38
|
Wehling-Henricks M, Tidball JG. Neuronal nitric oxide synthase-rescue of dystrophin/utrophin double knockout mice does not require nNOS localization to the cell membrane. PLoS One 2011; 6:e25071. [PMID: 22003386 PMCID: PMC3189177 DOI: 10.1371/journal.pone.0025071] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/26/2011] [Indexed: 02/03/2023] Open
Abstract
Survival of dystrophin/utrophin double-knockout (dko) mice was increased by muscle-specific expression of a neuronal nitric oxide synthase (nNOS) transgene. Dko mice expressing the transgene (nNOS TG+/dko) experienced delayed onset of mortality and increased life-span. The nNOS TG+/dko mice demonstrated a significant decrease in the concentration of CD163+, M2c macrophages that can express arginase and promote fibrosis. The decrease in M2c macrophages was associated with a significant reduction in fibrosis of heart, diaphragm and hindlimb muscles of nNOS TG+/dko mice. The nNOS transgene had no effect on the concentration of cytolytic, CD68+, M1 macrophages. Accordingly, we did not observe any change in the extent of muscle fiber lysis in the nNOS TG+/dko mice. These findings show that nNOS/NO (nitric oxide)-mediated decreases in M2c macrophages lead to a reduction in the muscle fibrosis that is associated with increased mortality in mice lacking dystrophin and utrophin. Interestingly, the dramatic and beneficial effects of the nNOS transgene were not attributable to localization of nNOS protein at the cell membrane. We did not detect any nNOS protein at the sarcolemma in nNOS TG+/dko muscles. This important observation shows that sarcolemmal localization is not necessary for nNOS to have beneficial effects in dystrophic tissue and the presence of nNOS in the cytosol of dystrophic muscle fibers can ameliorate the pathology and most importantly, significantly increase life-span.
Collapse
Affiliation(s)
- Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, United States of America
| | - James G. Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, United States of America
- Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, California, United States of America
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
39
|
The beta-isoform of neuronal nitric oxide synthase (nNOS) lacking the PDZ domain is localized at the sarcolemma. FEBS Lett 2011; 585:3219-23. [DOI: 10.1016/j.febslet.2011.09.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 09/09/2011] [Accepted: 09/12/2011] [Indexed: 11/21/2022]
|
40
|
Finanger Hedderick EL, Simmers JL, Soleimani A, Andres-Mateos E, Marx R, Files DC, King L, Crawford TO, Corse AM, Cohn RD. Loss of sarcolemmal nNOS is common in acquired and inherited neuromuscular disorders. Neurology 2011; 76:960-7. [PMID: 21403107 DOI: 10.1212/wnl.0b013e31821043c8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Neuronal nitric oxide synthase (nNOS), normally expressed at the sarcolemmal membrane, is known to be mislocalized to the sarcoplasm in several forms of muscular dystrophy. Our objectives were to characterize further the range of patients manifesting aberrant nNOS sarcolemmal immunolocalization and to study nNOS localization in animal models of nondystrophic myopathy. METHODS We carried out a retrospective cross-sectional study. We performed immunofluorescent staining for nNOS on biopsy specimens from 161 patients with acquired and nondystrophin inherited neuromuscular conditions. The localization of sarcolemmal nNOS correlated with mobility and functional status. Muscle specimens from mouse models of steroid-induced and starvation-related atrophy were studied for qualitative and quantitative nNOS expression. RESULTS Sarcolemmal nNOS staining was abnormal in 42% of patients with inherited myopathic conditions, 25% with acquired myopathic conditions, 57% with neurogenic conditions, and 93% with hypotonia. Interestingly, we found significant associations between mobility status or muscle function and sarcolemmal nNOS expression. Furthermore, mouse models of catabolic stress also demonstrated mislocalization of sarcolemmal nNOS. CONCLUSION Our analyses indicate that nNOS mislocalization is observed in a broad range of nondystrophic neuromuscular conditions associated with impaired mobility status and catabolic stress. Our findings suggest that the assessment of sarcolemmal localization of nNOS represents an important tool for the evaluation of muscle biopsies of patients with a variety of inherited and acquired forms of neuromuscular disorders.
Collapse
Affiliation(s)
- E L Finanger Hedderick
- Department of Neurology, Johns Hopkins University School of Medicine, 733 North Broadway BRB 529, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Huber-Abel FAM, Gerber M, Hoppeler H, Baum O. Exercise-induced angiogenesis correlates with the up-regulated expression of neuronal nitric oxide synthase (nNOS) in human skeletal muscle. Eur J Appl Physiol 2011; 112:155-62. [PMID: 21505843 DOI: 10.1007/s00421-011-1960-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 04/04/2011] [Indexed: 10/18/2022]
Abstract
The contribution of neuronal nitric oxide synthase (nNOS) to angiogenesis in human skeletal muscle after endurance exercise is controversially discussed. We therefore ascertained whether the expression of nNOS is associated with the capillary density in biopsies of the vastus lateralis (VL) muscle that had been derived from 10 sedentary male subjects before and after moderate training (four 30-min weekly jogging sessions for 6 months, with a heart-rate corresponding to 75% VO(2)max). In these biopsies, nNOS was predominantly expressed as alpha-isoform with exon-mu and to a lesser extent without exon-mu, as determined by RT-PCR. The mRNA levels of nNOS were quantified by real-time PCR and related to the capillary-to-fibre ratio and the numerical density of capillaries specified by light microscopy. If the VL biopsies of all subjects were co-analysed, mRNA levels of nNOS were non-significantly elevated after training (+34%; P > 0.05). However, only five of the ten subjects exhibited significant (P ≤ 0.05) elevations in the capillary-to-fibre ratio (+25%) and the numerical density of capillaries (+21%) and were thus undergoing angiogenesis. If the VL biopsies of these five subjects alone were evaluated, the mRNA levels of nNOS were significantly up-regulated (+128%; P ≤ 0.05) and correlated positively (r = 0.8; P ≤ 0.01) to angiogenesis. Accordingly, nNOS protein expression in VL biopsies quantified by immunoblotting was significantly increased (+82%; P ≤ 0.05) only in those subjects that underwent angiogenesis. In conclusion, the expression of nNOS at mRNA and protein levels was statistically linked to capillarity after exercise suggesting that nNOS is involved in the angiogenic response to training in human skeletal muscle.
Collapse
|
42
|
Gardan-Salmon D, Dixon JM, Lonergan SM, Selsby JT. Proteomic assessment of the acute phase of dystrophin deficiency in mdx mice. Eur J Appl Physiol 2011; 111:2763-73. [PMID: 21409400 DOI: 10.1007/s00421-011-1906-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Accepted: 03/03/2011] [Indexed: 11/27/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by the absence of a functional dystrophin protein and is modeled by the mdx mouse. The mdx mouse suffers an early necrotic bout in the hind limb muscles lasting from approximately 4 to 7 weeks. The purpose of this investigation was to determine the extent to which dystrophin deficiency changed the proteome very early in the disease process. In order to accomplish this, proteins from gastrocnemius from 6-week-old C57 (n = 6) and mdx (n = 6) mice were labeled with fluorescent dye and subjected to two-dimensional differential in-gel electrophoresis (2D-DIGE). Resulting differentially expressed spots were excised and protein identity determined via MALDI-TOF followed by database searching using MASCOT. Proteins of the immediate energy system and glycolysis were generally down-regulated in mdx mice compared to C57 mice. Conversely, expression of proteins involved in the Kreb's cycle and electron transport chain were increased in dystrophin-deficient muscle compared to control. Expression of cytoskeletal components, including tubulins, vimentin, and collagen, were increased in mdx mice compared to C57 mice. Importantly, these changes are occurring at only 6 weeks of age and are caused by acute dystrophin deficiency rather than more chronic injury. These data may provide insight regarding early pathologic changes occurring in dystrophin-deficient skeletal muscle.
Collapse
Affiliation(s)
- D Gardan-Salmon
- Department of Animal Science, Iowa State University, 2356 Kildee Hall, Ames, IA 50011, USA
| | | | | | | |
Collapse
|
43
|
Pegoraro E, Hoffman EP, Piva L, Gavassini BF, Cagnin S, Ermani M, Bello L, Soraru G, Pacchioni B, Bonifati MD, Lanfranchi G, Angelini C, Kesari A, Lee I, Gordish-Dressman H, Devaney JM, McDonald CM, Cooperative International Neuromuscular Research Group. SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy. Neurology 2011; 76:219-26. [PMID: 21178099 PMCID: PMC3034396 DOI: 10.1212/wnl.0b013e318207afeb] [Citation(s) in RCA: 173] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 07/27/2010] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is the most common single-gene lethal disorder. Substantial patient-patient variability in disease onset and progression and response to glucocorticoids is seen, suggesting genetic or environmental modifiers. METHODS Two DMD cohorts were used as test and validation groups to define genetic modifiers: a Padova longitudinal cohort (n = 106) and the Cooperative International Neuromuscular Research Group (CINRG) cross-sectional natural history cohort (n = 156). Single nucleotide polymorphisms to be genotyped were selected from mRNA profiling in patients with severe vs mild DMD, and genome-wide association studies in metabolism and polymorphisms influencing muscle phenotypes in normal volunteers were studied. RESULTS Effects on both disease progression and response to glucocorticoids were observed with polymorphism rs28357094 in the gene promoter of SPP1 (osteopontin). The G allele (dominant model; 35% of subjects) was associated with more rapid progression (Padova cohort log rank p = 0.003), and 12%-19% less grip strength (CINRG cohort p = 0.0003). CONCLUSIONS Osteopontin genotype is a genetic modifier of disease severity in Duchenne dystrophy. Inclusion of genotype data as a covariate or in inclusion criteria in DMD clinical trials would reduce intersubject variance, and increase sensitivity of the trials, particularly in older subjects.
Collapse
Affiliation(s)
- E Pegoraro
- Neuromuscular Center, Department of Neurosciences, University of Padova, 35128 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
T Abresch, E Henricson, M Cregan, M Glicke, J Han, L Johnson, N Joyce, C Chidambaranathan, S Kumar, D Biggar, L Eliasoph, V Harris, E Hosaki, J Mah, A Chiu, E Goia, L Walker, C Wright, M Yousefi, M Tulinius, A C Ahlander, L Berland, A K Kroksmark, U Sterky, D Escolar, R Leshner, M Birkmeier, S Kaminski, K Parker, C Spurney, C Tesi-Rocha, A Kornberg, K Carroll, K Devalle, R Kennedy, M Ryan, D Villano, Y Nevo, E Wisband, D Yaffe, A Dubrovsky, J Corderi, L Levy, L Mesa, N Kuntz, K Coleman, S Driscoll, A Hoffman, W Korn-Peterson, D Selcen, K Gorni, L Capone, P Clemens, H Abdel-Hamid, C Bise, A Craig, L Hache, C Nguyen, A Connolly, J Florence, B Malkus, A Pestronk, R Renna, J Schierbecker, C Siener, C Wulf, J Teasley, S Blair, B Grillo, E Monasterio, T Bertorini, J Clifft, C Feliciano, M Igarashi, K North, T Juarez, K Rose, R Webster, S Wicks, H Kolski, L Chen, C Kennedy, G Parks, J Wohlers, J Carlo, B Deliz, S Espada, P Fuste, C Luciano, T Lotze, H Farber, A Gupta, S Habetz, J Jeffries, R McNeil, J Day, A Erickson, M Margolis, G Parry, D Walk, A Cnaan, A Arrieta, N Bartley, P Canelos, T Duong, F Hu, A Zimmerman,
Collapse
|
44
|
Meadows E, Flynn JM, Klein WH. Myogenin regulates exercise capacity but is dispensable for skeletal muscle regeneration in adult mdx mice. PLoS One 2011; 6:e16184. [PMID: 21264243 PMCID: PMC3021523 DOI: 10.1371/journal.pone.0016184] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 12/15/2010] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most prevalent inherited childhood muscle disorder in humans. mdx mice exhibit a similar pathophysiology to the human disorder allowing for an in-depth investigation of DMD. Myogenin, a myogenic regulatory factor, is best known for its role in embryonic myogenesis, but its role in adult muscle maintenance and regeneration is still poorly understood. Here, we generated an mdx:Myogflox/flox mouse harboring a tamoxifen-inducible Cre recombinase transgene, which was used to conditionally delete Myog during adult life. After tamoxifen treatment, three groups of mice were created to study the effects of Myog deletion: mdx:Myogflox/flox mice (mdx), Myogflox/flox mice (wild-type), and mdx:MyogfloxΔ/floxΔ:Cre-ER mice (mdx:Myog-deleted). mdx:Myog-deleted mice exhibited no adverse phenotype and behaved normally. When run to exhaustion, mdx:Myog-deleted mice demonstrated an enhanced capacity for exercise compared to mdx mice, running nearly as far as wild-type mice. Moreover, these mice showed the same signature characteristics of muscle regeneration as mdx mice. Unexpectedly, we found that myogenin was dispensable for muscle regeneration. Factors associated with muscle fatigue, metabolism, and proteolysis were significantly altered in mdx:Myog-deleted mice, and this might contribute to their increased exercise capacity. Our results reveal novel functions for myogenin in adult muscle and suggest that reducing Myog expression in other muscle disease models may partially restore muscle function.
Collapse
Affiliation(s)
- Eric Meadows
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Graduate Training Program in Genes and Development, The University of Texas School of Biomedical Sciences at Houston, Houston, Texas, United States of America
| | - Jesse M. Flynn
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Graduate Training Program in Genes and Development, The University of Texas School of Biomedical Sciences at Houston, Houston, Texas, United States of America
| | - William H. Klein
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
45
|
Percival JM, Adamo CM, Beavo JA, Froehner SC. Evaluation of the therapeutic utility of phosphodiesterase 5A inhibition in the mdx mouse model of duchenne muscular dystrophy. Handb Exp Pharmacol 2011:323-44. [PMID: 21695647 DOI: 10.1007/978-3-642-17969-3_14] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating and ultimately fatal disease characterized by progressive muscle wasting and weakness. DMD is caused by the absence of a functional dystrophin protein, which in turn leads to reduced expression and mislocalization of dystrophin-associated proteins including neuronal nitric oxide (NO) synthase mu (nNOSμ). Disruption of nNOSμ signaling results in muscle fatigue and unopposed sympathetic vasoconstriction during exercise, thereby increasing contraction-induced damage in dystrophin-deficient muscles. The loss of normal nNOSμ signaling during exercise is central to the vascular dysfunction proposed over 40 years ago to be an important pathogenic mechanism in DMD. Recent preclinical studies focused on circumventing defective nNOSμ signaling in dystrophic skeletal and cardiac muscle by inhibiting phosphodiesterase 5A (PDE5A) have shown promising results. This review addresses nNOS signaling in normal and dystrophin-deficient muscles and the potential of PDE5A inhibition as a therapeutic approach for the treatment of cardiovascular deficits in DMD.
Collapse
Affiliation(s)
- Justin M Percival
- Department of Physiology and Biophysics, University of Washington, 357290, 98195-7290, Seattle, WA, USA.
| | | | | | | |
Collapse
|
46
|
Sciorati C, Buono R, Azzoni E, Casati S, Ciuffreda P, D'Angelo G, Cattaneo D, Brunelli S, Clementi E. Co-administration of ibuprofen and nitric oxide is an effective experimental therapy for muscular dystrophy, with immediate applicability to humans. Br J Pharmacol 2010; 160:1550-60. [PMID: 20590643 PMCID: PMC2938824 DOI: 10.1111/j.1476-5381.2010.00809.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/08/2010] [Accepted: 03/11/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Current therapies for muscular dystrophy are based on corticosteroids. Significant side effects associated with these therapies have prompted several studies aimed at identifying possible alternative strategies. As inflammation and defects of nitric oxide (NO) generation are key pathogenic events in muscular dystrophies, we have studied the effects of combining the NO donor isosorbide dinitrate (ISDN) and the non-steroidal anti-inflammatory drug ibuprofen. EXPERIMENTAL APPROACH alpha-Sarcoglycan-null mice were treated for up to 8 months with ISDN (30 mg.kg(-1)) plus ibuprofen (50 mg.kg(-1)) administered daily in the diet. Effects of ISDN and ibuprofen alone were assessed in parallel. Drug effects on animal motility and muscle function, muscle damage, inflammatory infiltrates and cytokine levels, as well as muscle regeneration including assessment of endogenous stem cell pool, were measured at selected time points. KEY RESULTS Combination of ibuprofen and ISDN stimulated regeneration capacity, of myogenic precursor cells, reduced muscle necrotic damage and inflammation. Muscle function in terms of free voluntary movement and resistance to exercise was maintained throughout the time window analysed. The effects of ISDN and ibuprofen administered separately were transient and significantly lower than those induced by their combination. CONCLUSIONS AND IMPLICATIONS Co-administration of NO and ibuprofen provided synergistic beneficial effects in a mouse model of muscular dystrophy, leading to an effective therapy. Our results open the possibility of immediate clinical testing of a combination of ISDN and ibuprofen in dystrophic patients, as both components are approved for use in humans, with a good safety profile.
Collapse
Affiliation(s)
- Clara Sciorati
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
| | - Roberta Buono
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Emanuele Azzoni
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Silvana Casati
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Pierangela Ciuffreda
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | | | - Dario Cattaneo
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Silvia Brunelli
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
- E. Medea Scientific InstituteBosisio Parini, Italy
| |
Collapse
|