1
|
Zhu GZ, Zhao K, Li HZ, Wu DZ, Chen YB, Han D, Gao JW, Chen XY, Yu YP, Huang ZW, Tu C, Zhong ZM. Melatonin ameliorates age-related sarcopenia by inhibiting fibrogenic conversion of satellite cell. Mol Med 2024; 30:238. [PMID: 39614149 DOI: 10.1186/s10020-024-00998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
The fibrogenic conversion of satellite cells contributes to the atrophy and fibrosis of skeletal muscle, playing a significant role in the pathogenesis of age-related sarcopenia. Melatonin, a hormone secreted by the pineal gland, exhibits anti-aging and anti-fibrotic effects in various conditions. However, the effect of melatonin on satellite cell fate and age-related sarcopenia remains under-explored. Here, we report that melatonin treatment mitigated the loss of muscle mass and strength in aged mice, replenished the satellite cell pool and curtailed muscle fibrosis. When primary SCs were cultured in vitro and subjected to aging induction via D-galactose, they exhibited a diminished myogenic potential and a conversion from myogenic to fibrogenic lineage. Notably, melatonin treatment effectively restored the myogenic potential and inhibited this lineage conversion. Furthermore, melatonin attenuated the expression of the fibrogenic cytokine, transforming growth factor-β1, and reduced the phosphorylation of its downstream targets Smad2/3 both in vivo and in vitro. In summary, our findings show melatonin's capacity to counteract muscle decline and inhibit fibrogenic conversion in aging SCs and highlight its potential therapeutic value for age-related sarcopenia.
Collapse
Affiliation(s)
- Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Zhao
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yong-Peng Yu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhi-Wei Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
2
|
Wei Y, Jiang Y, Lu Y, Hu Q. Histone modifications in Duchenne muscular dystrophy: pathogenesis insights and therapeutic implications. J Med Genet 2024; 61:1003-1010. [PMID: 39327039 DOI: 10.1136/jmg-2024-110045] [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: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a commonly encountered genetic ailment marked by loss-of-function mutations in the Dystrophin gene, ultimately resulting in progressive debilitation of skeletal muscle. The investigation into the pathogenesis of DMD has increasingly converged on the role of histone modifications within the broader context of epigenetic regulation. These modifications, including histone acetylation, methylation and phosphorylation, are catalysed by specific enzymes and play a critical role in gene expression. This article provides an overview of the histone modifications occurring in DMD and analyses the research progress and potential of different types of histone modifications in DMD due to changes in cellular signalling for muscle regeneration, to provide new insights into diagnostic and therapeutic options for DMD.
Collapse
Affiliation(s)
- Yanning Wei
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Key Laboratory of Biological Molecular Medicine Research of Education, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yufei Lu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Diseases, Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
3
|
Giri A, Mehan S, Khan Z, Das Gupta G, Narula AS, Kalfin R. Modulation of neural circuits by melatonin in neurodegenerative and neuropsychiatric disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3867-3895. [PMID: 38225412 DOI: 10.1007/s00210-023-02939-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/30/2023] [Indexed: 01/17/2024]
Abstract
Neurodegenerative and neuropsychiatric disorders are two broad categories of neurological disorders characterized by progressive impairments in movement and cognitive functions within the central and peripheral nervous systems, and have emerged as a significant cause of mortality. Oxidative stress, neuroinflammation, and neurotransmitter imbalances are recognized as prominent pathogenic factors contributing to cognitive deficits and neurobehavioral anomalies. Consequently, preventing neurodegenerative and neuropsychiatric diseases has surfaced as a pivotal challenge in contemporary public health. This review explores the investigation of neurodegenerative and neuropsychiatric disorders using both synthetic and natural bioactive compounds. A central focus lies on melatonin, a neuroregulatory hormone secreted by the pineal gland in response to light-dark cycles. Melatonin, an amphiphilic molecule, assumes multifaceted roles, including scavenging free radicals, modulating energy metabolism, and synchronizing circadian rhythms. Noteworthy for its robust antioxidant and antiapoptotic properties, melatonin exhibits diverse neuroprotective effects. The inherent attributes of melatonin position it as a potential key player in the pathophysiology of neurological disorders. Preclinical and clinical studies have demonstrated melatonin's efficacy in alleviating neuropathological symptoms across neurodegenerative and neuropsychiatric conditions (depression, schizophrenia, bipolar disorder, and autism spectrum disorder). The documented neuroprotective prowess of melatonin introduces novel therapeutic avenues for addressing neurodegenerative and psychiatric disorders. This comprehensive review encompasses many of melatonin's applications in treating diverse brain disorders. Despite the strides made, realizing melatonin's full neuroprotective potential necessitates further rigorous clinical investigations. By unravelling the extended neuroprotective benefits of melatonin, future studies promise to deepen our understanding and augment the therapeutic implications against neurological deficits.
Collapse
Affiliation(s)
- Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India.
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia, 1113, Bulgaria
- Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad, 2700, Bulgaria
| |
Collapse
|
4
|
Jallouli S, Ghroubi S, Sakka S, Ben Dhia I, Damak M, Yahia A, Driss T, Mhiri C, Elleuch MH, Hammouda O. Effects of a nighttime melatonin ingestion on dynamic postural balance and muscle strength the following morning in people living with multiple sclerosis: A preliminary study. Clin Neurol Neurosurg 2024; 238:108165. [PMID: 38428060 DOI: 10.1016/j.clineuro.2024.108165] [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: 04/24/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Decreased endogenous melatonin concentrations in people with multiple sclerosis (PwMS) are associated with fatigue and pain that impair postural balance and muscle strength. Melatonin ingestion had analgesic and anti-fatigue effects. However, the acute effect of exogenous melatonin on dynamic postural stability and muscle strength has not been studied yet in PwMS. This study aimed to investigate the safety and the efficacy of a nighttime melatonin intake on dynamic postural balance and lower-extremity muscle strength the following morning in PwMS. METHODS Fourteen PwMS (28.36 ± 6.81 years) were assessed (8 a.m.) pre- and post-acute intake of melatonin or placebo (6mg, 30 minutes before nocturnal bedtime). Evaluated parameters included dynamic postural balance (force platform), lower-extremity muscle strength [Five-Repetition Sit-To-Stand Test (5-STST)], hand dexterity (Nine-Hole Peg Test), nociceptive pain [Visual Analogue Scale (VAS)], neuropathic pain [Neuropathic Pain 4 Questions (DN4)], sleep quality and fatigue perception [Hooper Index (HI)]. RESULTS In the frontal plane, melatonin reduced the center of pressure (CoP) path length (CoPL), CoPL in the anteroposterior axis (CoPLY) and CoP sway area (CoPAr) compared with placebo by 7.56% (p=0.02, Cohens'd (d)=1.24), 19.27% (p<0.001, d=2.60) and 13.82% (p<0.001, d=2.02), respectively. Melatonin induced a higher decrease in these posturographic parameters compared with placebo in the sagittal plane [CoPL: 9.10% (p=0.005, d=1.02), CoPLY: 4.29% (p=0.025, d=1.07) and CoPAr: 7.45% (p=0.038, d=0.74)]. Melatonin decreased 5-STST duration as well as VAS, DN4, HI-fatigue and HI-sleep scores compared with placebo by 8.19% (p=0.008, d=1.19), 5.74% (p=0.04, d=0.82), 27.30% (p=0.023, d=0.98), 40.15% (p=0.044, d=0.85) and 30.16% (p=0.012, d=1.10), respectively. CONCLUSION This preliminary study, among PwMS, showed that acute melatonin ingestion was safe and efficient for improving dynamic postural stability and lower-extremity muscle strength probably through its analgesic and anti-fatigue effects.
Collapse
Affiliation(s)
- Sonda Jallouli
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia; High Institute of Sport and Physical Education of Sfax, University of Sfax, Tunisia.
| | - Sameh Ghroubi
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Salma Sakka
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Imen Ben Dhia
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia; High Institute of Sport and Physical Education of Sfax, University of Sfax, Tunisia
| | - Mariem Damak
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Abdelmoneem Yahia
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Tarak Driss
- LINP2, UFR STAPS, University of Paris Nanterre, Nanterre, France
| | - Chokri Mhiri
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease, LR12SP19, Habib Bourguiba University Hospital, University of Sfax, Tunisia
| | - Mohamed Habib Elleuch
- Research laboratory: Evaluation and Management of Musculoskeletal System Pathologies, LR20ES09, Faculty of Medicine, University of Sfax, Tunisia
| | - Omar Hammouda
- LINP2, UFR STAPS, University of Paris Nanterre, Nanterre, France; Research Laboratory, Molecular bases of Human Pathology, LR19ES13, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| |
Collapse
|
5
|
Ge X, Wang C, Yang G, Maimaiti D, Hou M, Liu H, Yang H, Chen X, Xu Y, He F. Enhancement of mitochondrial energy metabolism by melatonin promotes vascularized skeletal muscle regeneration in a volumetric muscle loss model. Free Radic Biol Med 2024; 210:146-157. [PMID: 38008130 DOI: 10.1016/j.freeradbiomed.2023.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/16/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
Volumetric muscle loss (VML) is a condition that results in the extensive loss of 20 % or more of skeletal muscle due to trauma or tumor ablation, leading to severe functional impairment and permanent disability. The current surgical interventions have limited functional regeneration of skeletal muscle due to the compromised self-repair mechanism. Melatonin has been reported to protect skeletal muscle from exercise-induced oxidative damage and holds great potential to treat muscle diseases. In this study, we hypothesize that melatonin can enhance myoblast differentiation and promote effective recovery of skeletal muscle following VML. In vitro administration of melatonin resulted in a significant enhancement of myogenesis in C2C12 myoblast cells, as evidenced by the up-regulation of myogenic marker genes in a dose-dependent manner. Further experiments revealed that silent information of regulator type 3 (SIRT3) played a critical role in the melatonin-enhanced myoblast differentiation through enhancement of mitochondrial energy metabolism and activation of mitochondrial antioxidant enzymes such as superoxide dismutase 2 (SOD2). Silencing of Sirt3 completely abrogated the protective effect of melatonin on the mitochondrial function of myoblasts, evidenced by the increased reactive oxygen species, decreased adenosine triphosphate production, and down-regulated myoblast-specific marker gene expression. In order to attain a protracted and consistent release, liposome-encapsuled melatonin was integrated into gelatin methacryloyl hydrogel (GelMA-Lipo@MT). The implantation of GelMA-Lipo@MT into a tibialis anterior muscle defect in a VML model effectively stimulated the formation of myofibers and new blood vessels in situ, while concurrently inhibiting fibrotic collagen deposition. The findings of this study indicate that the incorporation of melatonin with GelMA hydrogel has facilitated the de novo vascularized skeletal muscle regeneration by augmenting mitochondrial energy metabolism. This represents a promising approach for the development of skeletal muscle tissue engineering, which could be utilized for the treatment of VML and other severe muscle injuries.
Collapse
Affiliation(s)
- Xiaoyang Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Chengyue Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Guanyu Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Dimulati Maimaiti
- Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China; School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China; Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Hao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China
| | - Xi Chen
- School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China; Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China.
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, China.
| |
Collapse
|
6
|
Effects of melatonin ingestion on physical performance and biochemical responses following exhaustive running exercise in soccer players. Biol Sport 2022; 39:473-479. [PMID: 35309526 PMCID: PMC8919877 DOI: 10.5114/biolsport.2022.106385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 05/15/2021] [Indexed: 12/30/2022] Open
Abstract
Antioxidant supplementation has become a common practice among athletes to boost sport achievement. Likewise, melatonin (MEL) has been ingested as an ergogenic aid to improve physical performance. To date, no study has checked whether the multiple beneficial effects of MEL have an outcome during a maximum running exercise until exhaustion. Therefore, the present study aimed to evaluate the effect of MEL ingestion on physical performance and biochemical responses (i.e., oxidative stress) during exhaustive exercise. In a double blind randomized study, thirteen professional soccer players [age: 17.5 ± 0.8 years, body mass: 70.3 ± 3.9 kg, body height: 1.80 ± 0.08 m; maximal aerobic speed (MAS): 16.85 ± 0.63 km/h; mean ± standard deviation], members of a first league squad, performed a running exercise until exhaustion at 100% of MAS, after either MEL or placebo ingestion. Physical performance was assessed, and blood samples were obtained at rest and following the exercise. Compared to placebo, MEL intake prevented the increase in oxidative stress markers (i.e., malondialdehyde), alleviated the alteration of antioxidant status (i.e., glutathione peroxidase, uric acid and total bilirubin) and decreased post-exercise biomarkers of muscle damage (i.e., creatine kinase and lactate dehydrogenase) (p < 0.05). However, physical performance was not affected by MEL ingestion (p > 0.05). In conclusion, acute MEL intake before a maximal running exercise protected athletes from oxidative stress and cellular damage but without an effect on physical performance.
Collapse
|
7
|
Saad NY, Al-Kharsan M, Garwick-Coppens SE, Chermahini GA, Harper MA, Palo A, Boudreau RL, Harper SQ. Human miRNA miR-675 inhibits DUX4 expression and may be exploited as a potential treatment for Facioscapulohumeral muscular dystrophy. Nat Commun 2021; 12:7128. [PMID: 34880230 PMCID: PMC8654987 DOI: 10.1038/s41467-021-27430-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/18/2021] [Indexed: 01/02/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a potentially devastating myopathy caused by de-repression of the DUX4 gene in skeletal muscles. Effective therapies will likely involve DUX4 inhibition. RNA interference (RNAi) is one powerful approach to inhibit DUX4, and we previously described a RNAi gene therapy to achieve DUX4 silencing in FSHD cells and mice using engineered microRNAs. Here we report a strategy to direct RNAi against DUX4 using the natural microRNA miR-675, which is derived from the lncRNA H19. Human miR-675 inhibits DUX4 expression and associated outcomes in FSHD cell models. In addition, miR-675 delivery using gene therapy protects muscles from DUX4-associated death in mice. Finally, we show that three known miR-675-upregulating small molecules inhibit DUX4 and DUX4-activated FSHD biomarkers in FSHD patient-derived myotubes. To our knowledge, this is the first study demonstrating the use of small molecules to suppress a dominant disease gene using an RNAi mechanism.
Collapse
Affiliation(s)
- Nizar Y. Saad
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Mustafa Al-Kharsan
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA ,grid.266832.b0000 0001 2188 8502Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM USA
| | - Sara E. Garwick-Coppens
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Gholamhossein Amini Chermahini
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Madison A. Harper
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Andrew Palo
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Ryan L. Boudreau
- grid.214572.70000 0004 1936 8294Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA USA
| | - Scott Q. Harper
- grid.240344.50000 0004 0392 3476Center for Gene Therapy, the Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
8
|
Garegnani L, Hyland M, Roson Rodriguez P, Escobar Liquitay CM, Franco JV. Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy and progressive respiratory decline. Cochrane Database Syst Rev 2021; 12:CD013720. [PMID: 34850383 PMCID: PMC8632644 DOI: 10.1002/14651858.cd013720.pub3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder characterised by progressive muscle weakness beginning in early childhood. Respiratory failure and weak cough develop in all patients as a consequence of muscle weakness leading to a risk of atelectasis, pneumonia, or the need for ventilatory support. There is no curative treatment for DMD. Corticosteroids are the only pharmacological intervention proven to delay the onset and progression of muscle weakness and thus respiratory decline in DMD. Antioxidant treatment has been proposed to try to reduce muscle weakness in general, and respiratory decline in particular. OBJECTIVES: To assess the effects of antioxidant agents on preventing respiratory decline in people with Duchenne muscular dystrophy during the respiratory decline phase of the condition. SEARCH METHODS: We searched CENTRAL, MEDLINE, Embase, and two trials registers to 23 March 2021, together with reference checking, citation searching, and contact with study authors to identify additional studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs that met our inclusion criteria. We included male patients with a diagnosis of DMD who had respiratory decline evidenced by a forced vital capacity (FVC%) less than 80% but greater than 30% of predicted values, receiving any antioxidant agent compared with other therapies for the management of DMD or placebo. DATA COLLECTION AND ANALYSIS: Two review authors screened studies for eligibility, assessed risk of bias of studies, and extracted data. We used standard methods expected by Cochrane. We assessed the certainty of the evidence using the GRADE approach. The primary outcomes were FVC and hospitalisation due to respiratory infections. Secondary outcomes were quality of life, adverse events, change in muscle function, forced expiratory volume in the first second (FEV1), and peak expiratory flow (PEF). MAIN RESULTS: We included one study with 66 participants who were not co-treated with corticosteroids, which was the only study to contribute data to our main analysis. We also included a study that enrolled 255 participants treated with corticosteroids, which was only available as a press release without numerical results. The studies were parallel-group RCTs that assessed the effect of idebenone on respiratory function compared to placebo. The trial that contributed numerical data included patients with a mean (standard deviation) age of 14.3 (2.7) years at the time of inclusion, with a documented diagnosis of DMD or severe dystrophinopathy with clinical features consistent with typical DMD. The overall risk of bias across most outcomes was similar and judged as 'low'. Idebenone may result in a slightly less of a decline in FVC from baseline to one year compared to placebo (mean difference (MD) 3.28%, 95% confidence interval (CI) -0.41 to 6.97; 64 participants; low-certainty evidence), and probably has little or no effect on change in quality of life (MD -3.80, 95% CI -10.09 to 2.49; 63 participants; moderate-certainty evidence) (Pediatric Quality of Life Inventory (PedsQL), range 0 to 100, 0 = worst, 100 = best quality of life). As a related but secondary outcome, idebenone may result in less of a decline from baseline in FEV1 (MD 8.28%, 95% CI 0.89 to 15.67; 53 participants) and PEF (MD 6.27%, 95% CI 0.61 to 11.93; 1 trial, 64 participants) compared to placebo. Idebenone was associated with fewer serious adverse events (RR 0.42, 95% CI 0.09 to 2.04; 66 participants; low-certainty evidence) and little to no difference in non-serious adverse events (RR 1.00, 95% CI 0.88 to 1.13; 66 participants; low-certainty evidence) compared to placebo. Idebenone may result in little to no difference in change in arm muscle function (MD -2.45 N, 95% CI -8.60 to 3.70 for elbow flexors and MD -1.06 N, 95% CI -6.77 to 4.65 for elbow extensors; both 52 participants) compared to placebo. We found no studies evaluating the outcome hospitalisation due to respiratory infection. The second trial, involving 255 participants, for which data were available only as a press release without numerical data, was prematurely discontinued due to futility after an interim efficacy analysis based on FVC. There were no safety concerns. The certainty of the evidence was low for most outcomes due to imprecision and publication bias (the lack of a full report of the larger trial, which was prematurely terminated). AUTHORS' CONCLUSIONS Idebenone is the only antioxidant agent tested in RCTs for preventing respiratory decline in people with DMD for which evidence was available for assessment. Idebenone may result in slightly less of a decline in FVC and less of a decline in FEV1 and PEF, but probably has little to no measurable effect on change in quality of life. Idebenone is associated with fewer serious adverse events than placebo. Idebenone may result in little to no difference in change in muscle function. Discontinuation due to the futility of the SIDEROS trial and its expanded access programmes may indicate that idebenone research in this condition is no longer needed, but we await the trial data. Further research is needed to establish the effect of different antioxidant agents on preventing respiratory decline in people with DMD during the respiratory decline phase of the condition.
Collapse
Affiliation(s)
- Luis Garegnani
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Martin Hyland
- Paediatric Neurology Division - Paediatrics Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Roson Rodriguez
- Research Department, Instituto Universitario Hospital Italiano, Buenos Aires, Argentina
| | | | - Juan Va Franco
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
9
|
Garegnani L, Hyland M, Roson Rodriguez P, Escobar Liquitay CME, Franco JV. Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy and progressive respiratory decline. Cochrane Database Syst Rev 2021; 11:CD013720. [PMID: 34748221 PMCID: PMC8574769 DOI: 10.1002/14651858.cd013720.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder characterised by progressive muscle weakness beginning in early childhood. Respiratory failure and weak cough develop in all patients as a consequence of muscle weakness leading to a risk of atelectasis, pneumonia, or the need for ventilatory support. There is no curative treatment for DMD. Corticosteroids are the only pharmacological intervention proven to delay the onset and progression of muscle weakness and thus respiratory decline in DMD. Antioxidant treatment has been proposed to try to reduce muscle weakness in general, and respiratory decline in particular. OBJECTIVES: To assess the effects of antioxidant agents on preventing respiratory decline in people with Duchenne muscular dystrophy during the respiratory decline phase of the condition. SEARCH METHODS: We searched CENTRAL, MEDLINE, Embase, and two trials registers to 23 March 2021, together with reference checking, citation searching, and contact with study authors to identify additional studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs that met our inclusion criteria. We included male patients with a diagnosis of DMD who had respiratory decline evidenced by a forced vital capacity (FVC%) less than 80% but greater than 30% of predicted values, receiving any antioxidant agent compared with other therapies for the management of DMD or placebo. DATA COLLECTION AND ANALYSIS: Two review authors screened studies for eligibility, assessed risk of bias of studies, and extracted data. We used standard methods expected by Cochrane. We assessed the certainty of the evidence using the GRADE approach. The primary outcomes were FVC and hospitalisation due to respiratory infections. Secondary outcomes were quality of life, adverse events, change in muscle function, forced expiratory volume in the first second (FEV1), and peak expiratory flow (PEF). MAIN RESULTS: We included one study with 66 participants who were not co-treated with corticosteroids, which was the only study to contribute data to our main analysis. We also included a study that enrolled 255 participants treated with corticosteroids, which was only available as a press release without numerical results. The studies were parallel-group RCTs that assessed the effect of idebenone on respiratory function compared to placebo. The trial that contributed numerical data included patients with a mean (standard deviation) age of 14.3 (2.7) years at the time of inclusion, with a documented diagnosis of DMD or severe dystrophinopathy with clinical features consistent with typical DMD. The overall risk of bias across most outcomes was similar and judged as 'low'. Idebenone may result in a slightly less of a decline in FVC from baseline to one year compared to placebo (mean difference (MD) 3.28%, 95% confidence interval (CI) -0.41 to 6.97; 64 participants; low-certainty evidence), and probably has little or no effect on change in quality of life (MD -3.80, 95% CI -10.09 to 2.49; 63 participants; moderate-certainty evidence) (Pediatric Quality of Life Inventory (PedsQL), range 0 to 100, 0 = worst, 100 = best quality of life). As a related but secondary outcome, idebenone may result in less of a decline from baseline in FEV1 (MD 8.28%, 95% CI 0.89 to 15.67; 53 participants) and PEF (MD 6.27%, 95% CI 0.61 to 11.93; 1 trial, 64 participants) compared to placebo. Idebenone was associated with fewer serious adverse events (RR 0.42, 95% CI 0.09 to 2.04; 66 participants; low-certainty evidence) and little to no difference in non-serious adverse events (RR 1.00, 95% CI 0.88 to 1.13; 66 participants; low-certainty evidence) compared to placebo. Idebenone may result in little to no difference in change in arm muscle function (MD -2.45 N, 95% CI -8.60 to 3.70 for elbow flexors and MD -1.06 N, 95% CI -6.77 to 4.65 for elbow extensors; both 52 participants) compared to placebo. We found no studies evaluating the outcome hospitalisation due to respiratory infection. The second trial, involving 255 participants, for which data were available only as a press release without numerical data, was prematurely discontinued due to futility after an interim efficacy analysis based on FVC. There were no safety concerns. The certainty of the evidence was low for most outcomes due to imprecision and publication bias (the lack of a full report of the larger trial, which was prematurely terminated). AUTHORS' CONCLUSIONS Idebenone is the only antioxidant agent tested in RCTs for preventing respiratory decline in people with DMD for which evidence was available for assessment. Idebenone may result in slightly less of a decline in FVC and less of a decline in FEV1 and PEF, but probably has little to no measurable effect on change in quality of life. Idebenone is associated with fewer serious adverse events than placebo. Idebenone may result in little to no difference in change in muscle function. Discontinuation due to the futility of the SIDEROS trial and its expanded access programmes may indicate that idebenone research in this condition is no longer needed, but we await the trial data. Further research is needed to establish the effect of different antioxidant agents on preventing respiratory decline in people with DMD during the respiratory decline phase of the condition.
Collapse
Affiliation(s)
- Luis Garegnani
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Martin Hyland
- Paediatric Neurology Division - Paediatrics Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Roson Rodriguez
- Research Department, Instituto Universitario Hospital Italiano, Buenos Aires, Argentina
| | | | - Juan Va Franco
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
10
|
Wang YX, Yang GH, Zhang LL, Wang J, Wang JF. Melatonin as Immune Potentiator for Enhancing Subunit Vaccine Efficacy against Bovine Viral Diarrhea Virus. Vaccines (Basel) 2021; 9:vaccines9091039. [PMID: 34579276 PMCID: PMC8473004 DOI: 10.3390/vaccines9091039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) is a pathogen associated with substantial economic losses in the dairy cattle industry. Currently, there are no effective vaccines against BVDV. Melatonin (MT) has been shown to have anti-inflammatory and anti-viral properties, and the use of MF59 in vaccines significantly enhances vaccine efficiency. Here, MT and MF59 were added into the Erns-LTB vaccine. Subsequently, their inhibitory activity on the NF-κB signaling pathway in Mardin-Darby Bovine Kidney cells and the hippocampus was assessed using western blot and quantitative reverse transcription PCR. The findings revealed that MT in the Erns-LTB vaccine decreases the phosphorylation of p65 proteins caused by BVDV infection. In addition, MT decreased the mRNA levels of IL-1β and IL-6 in vitro, but increased the production of IFN-α, IFN-β, Mx1 in vitro, brain-derived neurotrophic factor, cyclic amp response element-binding protein, and the stem cell factor in vivo. Furthermore, treatment with Erns-LTB + MF59 + MT stimulated the production of T lymphocytes, alleviated pathological damage, decreased expressions of BVDV antigen, and tight junction proteins in mice. These findings imply that MT has potential for use in the Erns-LTB vaccine to inhibit BVDV infection and regulate the immune responses of T-cells by inhibiting the NF-κB signaling pathway.
Collapse
|
11
|
Sofela S, Sahloul S, Song YA. Biophysical analysis of drug efficacy on C. elegans models for neurodegenerative and neuromuscular diseases. PLoS One 2021; 16:e0246496. [PMID: 34115761 PMCID: PMC8195402 DOI: 10.1371/journal.pone.0246496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/25/2021] [Indexed: 11/25/2022] Open
Abstract
Caenorhabditis elegans has emerged as a powerful model organism for drug screening due to its cellular simplicity, genetic amenability and homology to humans combined with its small size and low cost. Currently, high-throughput drug screening assays are mostly based on image-based phenotyping with the focus on morphological-descriptive traits not exploiting key locomotory parameters of this multicellular model with muscles such as its thrashing force, a critical biophysical parameter when screening drugs for muscle-related diseases. In this study, we demonstrated the use of a micropillar-based force assay chip in combination with a fluorescence assay to evaluate the efficacy of various drugs currently used in treatment of neurodegenerative and neuromuscular diseases. Using this two-dimensional approach, we showed that the force assay was generally more sensitive in measuring efficacy of drug treatment in Duchenne Muscular Dystrophy and Parkinson's Disease mutant worms as well as partly in Amyotrophic Lateral Sclerosis model. These results underline the potential of our force assay chip in screening of potential drug candidates for the treatment of neurodegenerative and neuromuscular diseases when combined with a fluorescence assay in a two-dimensional analysis approach.
Collapse
Affiliation(s)
- Samuel Sofela
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Tandon School of Engineering, New York University, New York, NY, United States of America
| | - Sarah Sahloul
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Yong-Ak Song
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Tandon School of Engineering, New York University, New York, NY, United States of America
| |
Collapse
|
12
|
Ellwood RA, Piasecki M, Szewczyk NJ. Caenorhabditis elegans as a Model System for Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22094891. [PMID: 34063069 PMCID: PMC8125261 DOI: 10.3390/ijms22094891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
The nematode worm Caenorhabditis elegans has been used extensively to enhance our understanding of the human neuromuscular disorder Duchenne Muscular Dystrophy (DMD). With new arising clinically relevant models, technologies and treatments, there is a need to reconcile the literature and collate the key findings associated with this model.
Collapse
Affiliation(s)
- Rebecca A. Ellwood
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Mathew Piasecki
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence:
| |
Collapse
|
13
|
Jin H, Xie W, Hu P, Tang K, Wang X, Wu Y, He M, Yu D, Li Y. The role of melatonin in sarcopenia: Advances and application prospects. Exp Gerontol 2021; 149:111319. [PMID: 33753178 DOI: 10.1016/j.exger.2021.111319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022]
Abstract
Sarcopenia is an age-related disease that has gradually become a serious health problem for elderly individuals. It not only greatly increases the risk of falls, weakness, and disability but also reduces the ability of patients to take care of themselves. Sarcopenia can directly affect the quality of life and disease prognosis of elderly individuals. However, drug interventions for this disease are lacking. Melatonin is a biological hormone produced by the body that has good free radical scavenging effects, antioxidant effects and other effects. It was originally used as a sleep aid and is now being used for an increasing number of new indications. Its effect on sarcopenia has also begun to attract attention. It is currently known that it can protect the mitochondria of skeletal muscle cells, maintain the number of muscle fibres, partially reverse the pathological changes of ageing muscle tissue, and increase muscle strength in patients with sarcopenia. A large number of microRNAs are expressed during cell ageing, that in turn provides a biological background to age-related diseases, like sarcopenia. Increasing studies have found an interaction between melatonin and miRNAs, suggesting that melatonin can be used in the treatment of sarcopenia. The increased expression of inflammation-associated miRNA-483 in elderly patients may be the basis for the age-dependent decrease in melatonin secretion,that may play a role in the morbidity of sarcopenia. Melatonin is closely related to sarcopenia. It has a wide range of effects on sarcopenia and has good application prospects for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Hongfu Jin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Peiwu Hu
- Department of Scientific Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kun Tang
- Discipline Construction Office, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiuhua Wang
- Xiang Ya Nursing School, The Central South University, Changsha, China
| | - Yuxiang Wu
- School of Kinesiology, Jianghan University, Wuhan 430056, China
| | - Miao He
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Dengjie Yu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
14
|
Kopustinskiene DM, Bernatoniene J. Molecular Mechanisms of Melatonin-Mediated Cell Protection and Signaling in Health and Disease. Pharmaceutics 2021; 13:129. [PMID: 33498316 PMCID: PMC7909293 DOI: 10.3390/pharmaceutics13020129] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 01/07/2023] Open
Abstract
Melatonin, an endogenously synthesized indolamine, is a powerful antioxidant exerting beneficial action in many pathological conditions. Melatonin protects from oxidative stress in ischemic/reperfusion injury, neurodegenerative diseases, and aging, decreases inflammation, modulates the immune system, inhibits proliferation, counteracts the Warburg effect, and promotes apoptosis in various cancer models. Melatonin stimulates antioxidant enzymes in the cells, protects mitochondrial membrane phospholipids, especially cardiolipin, from oxidation thus preserving integrity of the membranes, affects mitochondrial membrane potential, stimulates activity of respiratory chain enzymes, and decreases the opening of mitochondrial permeability transition pore and cytochrome c release. This review will focus on the molecular mechanisms of melatonin effects in the cells during normal and pathological conditions and possible melatonin clinical applications.
Collapse
Affiliation(s)
- Dalia M. Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
| | - Jurga Bernatoniene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
| |
Collapse
|
15
|
Rugowska A, Starosta A, Konieczny P. Epigenetic modifications in muscle regeneration and progression of Duchenne muscular dystrophy. Clin Epigenetics 2021; 13:13. [PMID: 33468200 PMCID: PMC7814631 DOI: 10.1186/s13148-021-01001-z] [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: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a multisystemic disorder that affects 1:5000 boys. The severity of the phenotype varies dependent on the mutation site in the DMD gene and the resultant dystrophin expression profile. In skeletal muscle, dystrophin loss is associated with the disintegration of myofibers and their ineffective regeneration due to defective expansion and differentiation of the muscle stem cell pool. Some of these phenotypic alterations stem from the dystrophin absence-mediated serine-threonine protein kinase 2 (MARK2) misplacement/downregulation in activated muscle stem (satellite) cells and neuronal nitric oxide synthase loss in cells committed to myogenesis. Here, we trace changes in DNA methylation, histone modifications, and expression of regulatory noncoding RNAs during muscle regeneration, from the stage of satellite cells to myofibers. Furthermore, we describe the abrogation of these epigenetic regulatory processes due to changes in signal transduction in DMD and point to therapeutic treatments increasing the regenerative potential of diseased muscles based on this acquired knowledge.
Collapse
Affiliation(s)
- Anna Rugowska
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland
| | - Alicja Starosta
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland
| | - Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland.
| |
Collapse
|
16
|
Kourakis S, Timpani CA, de Haan JB, Gueven N, Fischer D, Rybalka E. Targeting Nrf2 for the treatment of Duchenne Muscular Dystrophy. Redox Biol 2021; 38:101803. [PMID: 33246292 PMCID: PMC7695875 DOI: 10.1016/j.redox.2020.101803] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
Imbalances in redox homeostasis can result in oxidative stress, which is implicated in various pathological conditions including the fatal neuromuscular disease Duchenne Muscular Dystrophy (DMD). DMD is a complicated disease, with many druggable targets at the cellular and molecular level including calcium-mediated muscle degeneration; mitochondrial dysfunction; oxidative stress; inflammation; insufficient muscle regeneration and dysregulated protein and organelle maintenance. Previous investigative therapeutics tended to isolate and focus on just one of these targets and, consequently, therapeutic activity has been limited. Nuclear erythroid 2-related factor 2 (Nrf2) is a transcription factor that upregulates many cytoprotective gene products in response to oxidants and other toxic stressors. Unlike other strategies, targeted Nrf2 activation has the potential to simultaneously modulate separate pathological features of DMD to amplify therapeutic benefits. Here, we review the literature providing theoretical context for targeting Nrf2 as a disease modifying treatment against DMD.
Collapse
Affiliation(s)
- Stephanie Kourakis
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia.
| | - Cara A Timpani
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia; Australian Institute for Musculoskeletal Science, Victoria University, St Albans, Victoria, Australia.
| | - Judy B de Haan
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia.
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia.
| | - Dirk Fischer
- Division of Developmental- and Neuropediatrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| | - Emma Rybalka
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia; Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia; Australian Institute for Musculoskeletal Science, Victoria University, St Albans, Victoria, Australia.
| |
Collapse
|
17
|
Peczkowski KK, Rastogi N, Lowe J, Floyd KT, Schultz EJ, Karaze T, Davis JP, Rafael-Fortney JA, Janssen PML. Muscle Twitch Kinetics Are Dependent on Muscle Group, Disease State, and Age in Duchenne Muscular Dystrophy Mouse Models. Front Physiol 2020; 11:568909. [PMID: 33101056 PMCID: PMC7545010 DOI: 10.3389/fphys.2020.568909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/28/2020] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disorder caused by the lack of functional dystrophin protein. In muscular dystrophy preclinical research, it is pertinent to analyze the force of the muscles affected by the disease to assess pathology and potential effectiveness of therapeutic interventions. Although muscles function at sub-maximal levels in vivo, maximal tetanic contractions are most commonly used to assess and report muscle function in muscular dystrophy studies. At submaximal activation, the kinetics of contraction and relaxation are heavily impacted by the kinetics of the single twitch. However, maximal tetanic force is often the main, if not sole, outcome measured in most studies, while contractile kinetics are rarely reported. To investigate the effect of muscle disease on twitch contraction kinetics, isolated diaphragm and extensor digitorum longus (EDL) muscles of 10-, 20-week, “het” (dystrophin deficient and utrophin haplo-insufficient), and 52-week mdx (dystrophin deficient) mice were analyzed and compared to wild-type controls. We observed that twitch contractile kinetics are dependent on muscle type, age, and disease state. Specific findings include that diaphragm from wildtype mice has a greater time to 50% relaxation (RT50) than time to peak tension (TTP) compared to the het and mdx dystrophic models, where there is a similar TTP compared to RT50. Diaphragm twitch kinetics remain virtually unchanged with age, while the EDL from het and mdx mice initially has a greater RT50 than TTP, but the TTP increases with age. The difference between EDL contractile kinetics of dystrophic and wildtype mice is more prominent at young age. Differences in kinetics yielded greater statistical significance compared to previously published force measurements, thus, using kinetics as an outcome parameter could potentially allow for use of smaller experimental groups in future study designs. Although this study focused on DMD models, our findings may be applicable to other skeletal muscle conditions and diseases.
Collapse
Affiliation(s)
- Kyra K Peczkowski
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Neha Rastogi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jeovanna Lowe
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kyle T Floyd
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Eric J Schultz
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Tallib Karaze
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
18
|
Garegnani L, Hyland M, Roson Rodriguez P, Escobar Liquitay CM, Quinlivan R, Franco JVA. Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy and progressive respiratory decline. Hippokratia 2020. [DOI: 10.1002/14651858.cd013720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Luis Garegnani
- Research Department; Instituto Universitario Hospital Italiano; Buenos Aires Argentina
| | - Martin Hyland
- Paediatric Neurology Division - Paediatrics Department; Hospital Italiano de Buenos Aires; Buenos Aires Argentina
| | - Pablo Roson Rodriguez
- Research Department; Instituto Universitario Hospital Italiano; Buenos Aires Argentina
| | | | - Rosaline Quinlivan
- MRC Centre for Neuromuscular Diseases and Dubowitz Neuromuscular Centre; UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery and Great Ormond Street; London UK
| | - Juan VA Franco
- Argentine Cochrane Centre; Instituto Universitario Hospital Italiano; Buenos Aires Argentina
| |
Collapse
|
19
|
Boccanegra B, Verhaart IEC, Cappellari O, Vroom E, De Luca A. Safety issues and harmful pharmacological interactions of nutritional supplements in Duchenne muscular dystrophy: considerations for Standard of Care and emerging virus outbreaks. Pharmacol Res 2020; 158:104917. [PMID: 32485610 PMCID: PMC7261230 DOI: 10.1016/j.phrs.2020.104917] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022]
Abstract
At the moment, little treatment options are available for Duchenne muscular dystrophy (DMD). The absence of the dystrophin protein leads to a complex cascade of pathogenic events in myofibres, including chronic inflammation and oxidative stress as well as altered metabolism. The attention towards dietary supplements in DMD is rapidly increasing, with the aim to counteract pathology-related alteration in nutrient intake, the consequences of catabolic distress or to enhance the immunological response of patients as nowadays for the COVID-19 pandemic emergency. By definition, supplements do not exert therapeutic actions, although a great confusion may arise in daily life by the improper distinction between supplements and therapeutic compounds. For most supplements, little research has been done and little evidence is available concerning their effects in DMD as well as their preventing actions against infections. Often these are not prescribed by clinicians and patients/caregivers do not discuss the use with their clinical team. Then, little is known about the real extent of supplement use in DMD patients. It is mistakenly assumed that, since compounds are of natural origin, if a supplement is not effective, it will also do no harm. However, supplements can have serious side effects and also have harmful interactions, in terms of reducing efficacy or leading to toxicity, with other therapies. It is therefore pivotal to shed light on this unclear scenario for the sake of patients. This review discusses the supplements mostly used by DMD patients, focusing on their potential toxicity, due to a variety of mechanisms including pharmacodynamic or pharmacokinetic interactions and contaminations, as well as on reports of adverse events. This overview underlines the need for caution in uncontrolled use of dietary supplements in fragile populations such as DMD patients. A culture of appropriate use has to be implemented between clinicians and patients' groups.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Ingrid E C Verhaart
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Duchenne Parent Project, the Netherlands
| | - Ornella Cappellari
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Elizabeth Vroom
- Duchenne Parent Project, the Netherlands; World Duchenne Organisation (UPPMD), the Netherlands
| | - Annamaria De Luca
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
20
|
Farjallah MA, Ghattassi K, Ben Mahmoud L, Graja A, Boudaya M, Elleuch H, Jammoussi K, Sahnoun Z, Souissi N, Chtourou H, Hammouda O. Effect of nocturnal melatonin intake on cellular damage and recovery from repeated sprint performance during an intensive training schedule. Chronobiol Int 2020; 37:686-698. [PMID: 32378422 DOI: 10.1080/07420528.2020.1746797] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
An optimal recovery between training sessions is of similar if not greater importance as the training content and program of the training, itself. One of the most used strategies for improving recovery is the ingestion of supplements. The present study aimed to evaluate the effect of 5 mg oral melatonin supplementation on the recovery from repeated sprint (RSA) of performance and biochemical responses (i.e. oxidative stress, leukocytosis cellular damage) after an intensive training camp (TC). Twenty soccer players performed an RSA test before and after an intensive six-day TC associated with nocturnal melatonin (n = 10) or placebo (n = 10) ingestion. Resting and post-RSA test blood samples were obtained before and after the TC. Compared to placebo, melatonin intake decreased resting oxidative stress markers (i.e, advanced oxidation protein products), leukocytosis (i.e. white blood cells (WBC), neutrophils (NE)) and biomarkers of cellular damage (i.e. creatine kinase (CK)). It also lowered post-exercise leukocytosis (i.e. WBC, NE, lymphocytes (LY), monocytes (MO)) and biomarkers of cellular damage (i.e. CK, aspartate aminotransferase (ASAT), alanine aminotransferase (ALAT)) and raised the activity of the main antioxidant enzymes (i.e. glutathione peroxidase (GPx), glutathione reductase (GR)). In addition, compared to placebo, melatonin reduced the deterioration of the best and total time during the RSA test after the TC. In conclusion, nocturnal melatonin supplementation during an intensive TC alleviated oxidative stress, leukocytosis and cellular damage and improved recovery of RSA performance in soccer players.
Collapse
Affiliation(s)
- Mohamed Amine Farjallah
- High Institute of Sport and Physical Education, Manouba University, Ksar-Saïd , Tunis, Tunisia
| | - Kais Ghattassi
- High Institute of Sport and Physical Education, Manouba University, Ksar-Saïd , Tunis, Tunisia
| | - Lobna Ben Mahmoud
- Department of Pharmacology, Faculty of Medicine, University of Sfax , Sfax, Tunisia
| | - Ahmed Graja
- High Institute of Sport and Physical Education, Manouba University, Ksar-Saïd , Tunis, Tunisia.,Research Laboratory, Education, Motricity, Sport and Health, LR15JS01, High Institute of Sport and Physical Education, University of Sfax , Sfax, Tunisia
| | - Mariem Boudaya
- Biochemistry Laboratory, CHU Hedi Chaker, University of Sfax , Sfax, Tunisia
| | - Henda Elleuch
- Hematology Laboratory, CHU Hedi Chaker, University of Sfax , Sfax, Tunisia
| | - Kamel Jammoussi
- Biochemistry Laboratory, CHU Hedi Chaker, University of Sfax , Sfax, Tunisia
| | - Zouheir Sahnoun
- Department of Pharmacology, Faculty of Medicine, University of Sfax , Sfax, Tunisia
| | - Nizar Souissi
- Physical Activity, Sport and Health, UR18JS01, National Observatory of Sport , Tunis, Tunisia
| | - Hamdi Chtourou
- Physical Activity, Sport and Health, UR18JS01, National Observatory of Sport , Tunis, Tunisia.,High Institute of Sport and Physical Education of Sfax, University of Sfax , Sfax, Tunisia
| | - Omar Hammouda
- Interdisciplinary Laboratory in Neurosciences, Physiology and Psychology: Physical Activity, Health and Learning (LINP2-APSA), UPL, Paris Nanterre University, UFR STAPS , Nanterre, France.,Research Laboratory, Molecular Bases of Human Pathology, LR12ES17, Faculty of Medicine, University of Sfax , Sfax, Tunisia
| |
Collapse
|
21
|
The regulatory role of melatonin in skeletal muscle. J Muscle Res Cell Motil 2020; 41:191-198. [PMID: 32157560 DOI: 10.1007/s10974-020-09578-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/22/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022]
Abstract
Melatonin (N-acetyl-5-methoxy-tryptamine) is an effective antioxidant and free radical scavenger, that has important biological effects in multiple cell types and species. Melatonin research in muscle has recently gained attention, mainly focused on its role in cells or tissue repair and regeneration after injury, due to its powerful biological functions, including its antioxidant, anti-inflammation, anti-tumor and anti-cancer, circadian rhythm, and anti-apoptotic effects. However, the effect of melatonin in regulating muscle development has not been systematically summarized. In this review, we outline the latest research on the involvement of melatonin in the regulation of muscle development and regeneration in order to better understand its underlying molecular mechanisms and potential applications.
Collapse
|
22
|
Stacchiotti A, Favero G, Rodella LF. Impact of Melatonin on Skeletal Muscle and Exercise. Cells 2020; 9:cells9020288. [PMID: 31991655 PMCID: PMC7072499 DOI: 10.3390/cells9020288] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle disorders are dramatically increasing with human aging with enormous sanitary costs and impact on the quality of life. Preventive and therapeutic tools to limit onset and progression of muscle frailty include nutrition and physical training. Melatonin, the indole produced at nighttime in pineal and extra-pineal sites in mammalians, has recognized anti-aging, anti-inflammatory, and anti-oxidant properties. Mitochondria are the favorite target of melatonin, which maintains them efficiently, scavenging free radicals and reducing oxidative damage. Here, we discuss the most recent evidence of dietary melatonin efficacy in age-related skeletal muscle disorders in cellular, preclinical, and clinical studies. Furthermore, we analyze the emerging impact of melatonin on physical activity. Finally, we consider the newest evidence of the gut-muscle axis and the influence of exercise and probably melatonin on the microbiota. In our opinion, this review reinforces the relevance of melatonin as a safe nutraceutical that limits skeletal muscle frailty and prolongs physical performance.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Correspondence: ; Tel.: +39-030-3717478; Fax: +39-030-3717486
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
| | - Luigi Fabrizio Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
23
|
Chen B, You W, Shan T. Myomaker, and Myomixer-Myomerger-Minion modulate the efficiency of skeletal muscle development with melatonin supplementation through Wnt/β-catenin pathway. Exp Cell Res 2019; 385:111705. [PMID: 31682812 DOI: 10.1016/j.yexcr.2019.111705] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/30/2019] [Accepted: 10/29/2019] [Indexed: 02/03/2023]
Abstract
Melatonin, a pleiotropic hormone secreted from the pineal gland, has been shown to exert beneficial effects in muscle regeneration and repair due to its functional diversity, including anti-inflammation, anti-apoptosis, and anti-oxidative activity. However, little is known about the negative role of melatonin in myogenesis. Here, using skeletal muscle cells, we found that melatonin promoted C2C12 cells proliferation and inhibits differentiation both in C2C12 cells and primary myoblasts in mice. Melatonin administration significantly down-regulated differentiation and fusion related genes and inhibited myotube formation both in C2C12 cells and primary myoblasts in mice. RNA-seq showed that melatonin down-regulated essential fusion pore components Myomaker and Myomixer-Myomerger-Minion. Moreover, melatonin suppressed Wnt/β-catenin signaling. Inhibition of GSK3β by LiCl rescued the influence of melatonin on differentiation efficiency, Myomaker, but not Myomxier in C2C12 cells. In conclusion, melatonin inhibits myogenic differentiation, Myomaker, and Myomixer through reducing Wnt/β-catenin signaling. These data establish a link between melatonin and fusogenic membrane proteins Myomaker and Myomixer, and suggest the new perspective of melatonin in treatment or preventment of muscular diseases.
Collapse
Affiliation(s)
- Bide Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.
| |
Collapse
|
24
|
Bjørklund G, Chirumbolo S, Dadar M, Pen JJ, Doşa MD, Pivina L, Semenova Y, Aaseth J. Insights on Nutrients as Analgesics in Chronic Pain. Curr Med Chem 2019; 27:6407-6423. [PMID: 31309880 DOI: 10.2174/0929867326666190712172015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 02/07/2023]
Abstract
Many serious inflammatory disorders and nutrient deficiencies induce chronic pain, and anti-inflammatory diets have been applied successfully to modify the inflammatory symptoms causing chronic pain. Numerous scientific data and clinical investigations have demonstrated that long-term inflammation could lead to an inappropriate or exaggerated sensibility to pain. In addition, some Non-steroidal Anti-inflammatory Drugs (NSAID), which directly act on the many enzymes involved in pain and inflammation, including cyclooxygenases, are used to dampen the algesic signal to the central nervous system, reducing the responses of soft C-fibers to pain stimuli. On the other hand, there are a few reports from both health authorities and physicians, reporting that decreased transmission of pain signals can be achieved and improved, depending on the patient's dietary habit. Many nutrients, as well as a suitable level of exercise (resistance training), are the best methods for improving the total mitochondrial capacity in muscle cells, which can lead to a reduction in sensitivity to pain, particularly by lowering the inflammatory signaling to C-fibers. According to the current literature, it could be proposed that chronic pain results from the changed ratio of neuropeptides, hormones, and poor nutritional status, often related to an underlying inflammatory disorder. The current review also evaluates the effective role of nutrition-related interventions on the severity of chronic pain. This review pointed out that nutritional interventions can have a positive effect on pain experience through the indirect inhibitory effect on prostaglandin E2 and attenuation of mitochondrial dysfunction caused by ischemia/reperfusion in skeletal muscle, improving the intracellular antioxidant defense system. These data highlight the need for more nutrition studies where chronic pain is the primary outcome, using accurate interventions. To date, no nutritional recommendation for chronic pain has been officially proposed. Therefore, the goal of this article is to explore pain management and pain modulation, searching for a mode of nutrition efficient in reducing pain.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy,CONEM Scientific Secretary, Verona, Italy
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Joeri J Pen
- Diabetes Clinic, Department of Internal Medicine, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium,Department of Nutrition, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Lyudmila Pivina
- Semey Medical University, Semey, Kazakhstan,CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, Semey, Kazakhstan
| | - Yulia Semenova
- Semey Medical University, Semey, Kazakhstan,CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, Semey, Kazakhstan
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway,Faculty of Health and Social Science, Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
25
|
Debruin DA, Andreacchio N, Hanson ED, Timpani CA, Rybalka E, Hayes A. The Effect of Vitamin D Supplementation on Skeletal Muscle in the mdx Mouse Model of Duchenne Muscular Dystrophy. Sports (Basel) 2019; 7:sports7050096. [PMID: 31035483 PMCID: PMC6572350 DOI: 10.3390/sports7050096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/29/2022] Open
Abstract
Vitamin D (VitD) has shown to be beneficial in reversing muscle weakness and atrophy associated with VitD deficiency. Duchenne muscular dystrophy is characterized by worsening muscle weakness and muscle atrophy, with VitD deficiency commonly observed. This study aimed to investigate the effect of VitD supplementation on dystrophic skeletal muscle. Eight-week old female control (C57BL/10; n = 29) and dystrophic (C57BL/mdx; n = 23) mice were randomly supplemented with one of three VitD enriched diets (1000, 8000 & 20,000 IU/kg chow). Following a four-week feeding period, the extensor digitorum longus (EDL) and soleus muscles contractile and fatigue properties were tested ex vivo, followed by histological analysis. As expected, mdx muscles displayed higher mass yet lower specific forces and a rightward shift in their force frequency relationship consistent with dystrophic pathology. There was a trend for mdx muscle mass to be larger following the 20,000 IU/kg diet, but this did not result in improved force production. Fiber area in the EDL was larger in mdx compared to controls, and there were higher amounts of damage in both muscles, with VitD supplementation having no effect. Four weeks of VitD supplementation did not appear to have any impact upon dystrophic skeletal muscle pathology at this age.
Collapse
Affiliation(s)
- Danielle A Debruin
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
- Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia.
| | - Nicola Andreacchio
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
| | - Erik D Hanson
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Cara A Timpani
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
- Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia.
| | - Emma Rybalka
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
- Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia.
| | - Alan Hayes
- Institute of Sport and Health, Victoria University, Melbourne 3011, Australia.
- Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia.
- Melbourne Medical School, The University of Melbourne, Melbourne 3010, Australia.
| |
Collapse
|
26
|
Rondanelli M, Peroni G, Gasparri C, Infantino V, Nichetti M, Cuzzoni G, Spadaccini D, Perna S. Is a Combination of Melatonin and Amino Acids Useful to Sarcopenic Elderly Patients? A Randomized Trial. Geriatrics (Basel) 2018; 4:geriatrics4010004. [PMID: 31023972 PMCID: PMC6473373 DOI: 10.3390/geriatrics4010004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
This study evaluated the effectiveness of a 4-week intervention of melatonin and essential aminoacid supplementation on body composition, protein metabolism, strength and inflammation in 159 elderly sarcopenic patients (42/117, men/women), assigned to four groups: isocaloric placebo (P, n = 44), melatonin (M, 1 mg/daily, n = 42), essential amino acids (eAA 4 g/daily, n = 40) or eAA plus melatonin (eAAM, 4 g eAA and 1 mg melatonin/daily, n = 30). Data from body composition (dual X-ray absortiometry (DXA)), strength (handgrip test) and biochemical parameters for the assessment of protein metabolism (albumin) and inflammation (CRP) were collected at baseline and after the 4-week intervention. Compared with P and M, supplementation with eAA plus M increased total fat-free mass (vs. P: +2190 g; p < 0.01; vs. M: +2107 g; p < 0.05). M alone lowered albumin levels (vs. P: −0.39 g; p < 0.01; vs. eAA: −0.47 g; p < 0.01). This data on albumin was confirmed by within-group analysis (M −0.44g; p < 0.001; eAAM: −0.34 p < 0.05). M and eAA seemed to lower the percentage of gynoid fat (p < 0.05) and android fat (p < 0.01). No significant changes in inflammation or strength were reported. A 4-week intervention with eAA plus M together may be effective in enhancing fat-free-mass compared to M and P but not versus eAA. M alone demonstrates a negative effect on albumin level.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- IRCCS Mondino Foundation, 27100 Pavia, Italy.
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
| | - Gabriella Peroni
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
| | - Clara Gasparri
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
| | - Vittoria Infantino
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
- Department of Biomedical Science and Human Oncology, University of Bari, 70121 Bari, Italy.
| | - Mara Nichetti
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
| | | | - Daniele Spadaccini
- Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, 27100 Pavia, Italy.
| | - Simone Perna
- Department of Biology, College of Science, University of Bahrain, Sakhir Campus P.O. Box 32038, Kingdom of Bahrain.
| |
Collapse
|
27
|
Hewitt JE, Pollard AK, Lesanpezeshki L, Deane CS, Gaffney CJ, Etheridge T, Szewczyk NJ, Vanapalli SA. Muscle strength deficiency and mitochondrial dysfunction in a muscular dystrophy model of Caenorhabditis elegans and its functional response to drugs. Dis Model Mech 2018; 11:dmm036137. [PMID: 30396907 PMCID: PMC6307913 DOI: 10.1242/dmm.036137] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022] Open
Abstract
Muscle strength is a key clinical parameter used to monitor the progression of human muscular dystrophies, including Duchenne and Becker muscular dystrophies. Although Caenorhabditis elegans is an established genetic model for studying the mechanisms and treatments of muscular dystrophies, analogous strength-based measurements in this disease model are lacking. Here, we describe the first demonstration of the direct measurement of muscular strength in dystrophin-deficient C. elegans mutants using a micropillar-based force measurement system called NemaFlex. We show that dys-1(eg33) mutants, but not dys-1(cx18) mutants, are significantly weaker than their wild-type counterparts in early adulthood, cannot thrash in liquid at wild-type rates, display mitochondrial network fragmentation in the body wall muscles, and have an abnormally high baseline mitochondrial respiration. Furthermore, treatment with prednisone, the standard treatment for muscular dystrophy in humans, and melatonin both improve muscular strength, thrashing rate and mitochondrial network integrity in dys-1(eg33), and prednisone treatment also returns baseline respiration to normal levels. Thus, our results demonstrate that the dys-1(eg33) strain is more clinically relevant than dys-1(cx18) for muscular dystrophy studies in C. elegans This finding, in combination with the novel NemaFlex platform, can be used as an efficient workflow for identifying candidate compounds that can improve strength in the C. elegans muscular dystrophy model. Our study also lays the foundation for further probing of the mechanism of muscle function loss in dystrophin-deficient C. elegans, leading to knowledge translatable to human muscular dystrophy.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jennifer E Hewitt
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Amelia K Pollard
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham & National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Leila Lesanpezeshki
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Colleen S Deane
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
| | - Christopher J Gaffney
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
- Lancaster Medical School, Furness College, Lancaster University, Lancaster LA1 4YG, UK
| | - Timothy Etheridge
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
| | - Nathaniel J Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham & National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
28
|
Gayi E, Neff LA, Massana Muñoz X, Ismail HM, Sierra M, Mercier T, Décosterd LA, Laporte J, Cowling BS, Dorchies OM, Scapozza L. Tamoxifen prolongs survival and alleviates symptoms in mice with fatal X-linked myotubular myopathy. Nat Commun 2018; 9:4848. [PMID: 30451843 PMCID: PMC6243013 DOI: 10.1038/s41467-018-07058-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/12/2018] [Indexed: 11/08/2022] Open
Abstract
X-linked myotubular myopathy (XLMTM, also known as XLCNM) is a severe congenital muscular disorder due to mutations in the myotubularin gene, MTM1. It is characterized by generalized hypotonia, leading to neonatal death of most patients. No specific treatment exists. Here, we show that tamoxifen, a well-known drug used against breast cancer, rescues the phenotype of Mtm1-deficient mice. Tamoxifen increases lifespan several-fold while improving overall motor function and preventing disease progression including lower limb paralysis. Tamoxifen corrects functional, histological and molecular hallmarks of XLMTM, with improved force output, myonuclei positioning, myofibrillar structure, triad number, and excitation-contraction coupling. Tamoxifen normalizes the expression level of the XLMTM disease modifiers DNM2 and PI3KC2B, likely contributing to the phenotypic rescue. Our findings demonstrate that tamoxifen is a promising candidate for clinical evaluation in XLMTM patients.
Collapse
MESH Headings
- Animals
- Class II Phosphatidylinositol 3-Kinases/genetics
- Class II Phosphatidylinositol 3-Kinases/metabolism
- Disease Models, Animal
- Disease Progression
- Dynamin II/genetics
- Dynamin II/metabolism
- Electric Stimulation
- Excitation Contraction Coupling/drug effects
- Female
- Gene Expression/drug effects
- Genes, Lethal
- Humans
- Longevity/drug effects
- Male
- Mice
- Mice, Knockout
- Motor Activity/drug effects
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myofibrils/drug effects
- Myofibrils/metabolism
- Myofibrils/ultrastructure
- Myopathies, Structural, Congenital/drug therapy
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/pathology
- Protective Agents/pharmacology
- Protein Tyrosine Phosphatases, Non-Receptor/deficiency
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Tamoxifen/pharmacology
Collapse
Affiliation(s)
- Elinam Gayi
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland
| | - Laurence A Neff
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland
| | - Xènia Massana Muñoz
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, 67404, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, 67404, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, 67404, France
- Université de Strasbourg, Illkirch, 67404, France
| | - Hesham M Ismail
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland
| | - Marta Sierra
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland
| | - Thomas Mercier
- Division and Laboratory of Clinical Pharmacology, Service of Biomedicine, Department of Laboratories, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Laurent A Décosterd
- Division and Laboratory of Clinical Pharmacology, Service of Biomedicine, Department of Laboratories, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, 67404, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, 67404, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, 67404, France
- Université de Strasbourg, Illkirch, 67404, France
| | - Belinda S Cowling
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, 67404, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, 67404, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, 67404, France
- Université de Strasbourg, Illkirch, 67404, France
| | - Olivier M Dorchies
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland.
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Lausanne, University of Geneva, CMU 5-6, Rue Michel-Servet 1, Geneva, 1211, Switzerland.
| |
Collapse
|
29
|
Nutrition in Duchenne muscular dystrophy 16–18 March 2018, Zaandam, the Netherlands. Neuromuscul Disord 2018; 28:680-689. [DOI: 10.1016/j.nmd.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 11/17/2022]
|
30
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current and emerging therapies for Duchenne muscular dystrophy (DMD). RECENT FINDINGS Coinciding with new standardized care guidelines, there are a growing number of therapeutic options to treat males with DMD. Treatment of the underlying pathobiology, such as micro-dystrophin gene replacement, exon skipping, stop codon read-through agents, and utrophin modulators showed variable success in animal and human studies. Symptomatic therapies to target muscle ischemia, enhance muscle regeneration, prevent muscle fibrosis, inhibit myostatin, and reduce inflammation are also under investigation. DMD is a complex, heterogeneous degenerative disease. The pharmacological and technological achievements made in recent years, plus timely supportive interventions will likely lead to an improved quality of life for many individuals with DMD.
Collapse
Affiliation(s)
- Megan Crone
- Division of Neurology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. .,Alberta Children's Hospital, 2888 Shaganappi Trail NW, Calgary, Alberta, T3B 6A8, Canada.
| | - Jean K Mah
- Division of Neurology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Ismail HM, Dorchies OM, Scapozza L. The potential and benefits of repurposing existing drugs to treat rare muscular dystrophies. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1452733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hesham M. Ismail
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Olivier M. Dorchies
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| |
Collapse
|
32
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
33
|
Meng X, Li Y, Li S, Zhou Y, Gan RY, Xu DP, Li HB. Dietary Sources and Bioactivities of Melatonin. Nutrients 2017; 9:E367. [PMID: 28387721 PMCID: PMC5409706 DOI: 10.3390/nu9040367] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/14/2017] [Accepted: 03/31/2017] [Indexed: 12/14/2022] Open
Abstract
Insomnia is a serious worldwide health threat, affecting nearly one third of the general population. Melatonin has been reported to improve sleep efficiency and it was found that eating melatonin-rich foods could assist sleep. During the last decades, melatonin has been widely identified and qualified in various foods from fungi to animals and plants. Eggs and fish are higher melatonin-containing food groups in animal foods, whereas in plant foods, nuts are with the highest content of melatonin. Some kinds of mushrooms, cereals and germinated legumes or seeds are also good dietary sources of melatonin. It has been proved that the melatonin concentration in human serum could significantly increase after the consumption of melatonin containing food. Furthermore, studies show that melatonin exhibits many bioactivities, such as antioxidant activity, anti-inflammatory characteristics, boosting immunity, anticancer activity, cardiovascular protection, anti-diabetic, anti-obese, neuroprotective and anti-aging activity. This review summaries the dietary sources and bioactivities of melatonin, with special attention paid to the mechanisms of action.
Collapse
Affiliation(s)
- Xiao Meng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ya Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China.
| | - Yue Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ren-You Gan
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China.
| | - Dong-Ping Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
- South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
34
|
Woodman KG, Coles CA, Lamandé SR, White JD. Nutraceuticals and Their Potential to Treat Duchenne Muscular Dystrophy: Separating the Credible from the Conjecture. Nutrients 2016; 8:E713. [PMID: 27834844 PMCID: PMC5133099 DOI: 10.3390/nu8110713] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/20/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
In recent years, complementary and alternative medicine has become increasingly popular. This trend has not escaped the Duchenne Muscular Dystrophy community with one study showing that 80% of caregivers have provided their Duchenne patients with complementary and alternative medicine in conjunction with their traditional treatments. These statistics are concerning given that many supplements are taken based on purely "anecdotal" evidence. Many nutraceuticals are thought to have anti-inflammatory or anti-oxidant effects. Given that dystrophic pathology is exacerbated by inflammation and oxidative stress these nutraceuticals could have some therapeutic benefit for Duchenne Muscular Dystrophy (DMD). This review gathers and evaluates the peer-reviewed scientific studies that have used nutraceuticals in clinical or pre-clinical trials for DMD and thus separates the credible from the conjecture.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/adverse effects
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antioxidants/adverse effects
- Antioxidants/therapeutic use
- Biomedical Research/methods
- Biomedical Research/trends
- Combined Modality Therapy/adverse effects
- Dietary Supplements/adverse effects
- Evidence-Based Medicine
- Humans
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscular Dystrophy, Duchenne/diet therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/therapy
- Peer Review, Research/methods
- Peer Review, Research/trends
- Reproducibility of Results
- Severity of Illness Index
Collapse
Affiliation(s)
- Keryn G Woodman
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Australia.
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville 3010, Australia.
| | - Chantal A Coles
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Australia.
| | - Shireen R Lamandé
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Australia.
- Department of Pediatrics, The University of Melbourne, Parkville 3010, Australia.
| | - Jason D White
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Australia.
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
35
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
36
|
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy in childhood. It is caused by mutations of the DMD gene, leading to progressive muscle weakness, loss of independent ambulation by early teens, and premature death due to cardiorespiratory complications. The diagnosis can usually be made after careful review of the history and examination of affected boys presenting with developmental delay, proximal weakness, and elevated serum creatine kinase, plus confirmation by muscle biopsy or genetic testing. Precise characterization of the DMD mutation is important for genetic counseling and individualized treatment. Current standard of care includes the use of corticosteroids to prolong ambulation and to delay the onset of secondary complications. Early use of cardioprotective agents, noninvasive positive pressure ventilation, and other supportive strategies has improved the life expectancy and health-related quality of life for many young adults with DMD. New emerging treatment includes viral-mediated microdystrophin gene replacement, exon skipping to restore the reading frame, and nonsense suppression therapy to allow translation and production of a modified dystrophin protein. Other potential therapeutic targets involve upregulation of compensatory proteins, reduction of the inflammatory cascade, and enhancement of muscle regeneration. So far, data from DMD clinical trials have shown limited success in delaying disease progression; unforeseen obstacles included immune response against the generated mini-dystrophin, inconsistent evidence of dystrophin production in muscle biopsies, and failure to demonstrate a significant improvement in the primary outcome measure, as defined by the 6-minute walk test in some studies. The long-term safety and efficacy of emerging treatments will depend on the selection of appropriate clinical end points and sensitive biomarkers to detect meaningful changes in disease progression. Correction of the underlying mutations using new gene-editing technologies and corticosteroid analogs with better safety profiles offers renewed hope for many individuals with DMD and their families.
Collapse
Affiliation(s)
- Jean K Mah
- Department of Pediatrics and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
37
|
Retracted: 'The effects of Duchenne muscular dystrophy on the performance of the stomatognathic system: case-control study', by Ferreira B., Da Silva G.P., Gonçalves C.R., et al. Dev Med Child Neurol 2016; 58:523. [PMID: 25683793 DOI: 10.1111/dmcn.12707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The above article, published online on 13 February 2015 in Wiley Online Library Early View (wileyonlinelibrary.com), has been retracted by agreement between the authors, the journal editors, Bernard Dan and Peter Rosenbaum, the MacKeith Press and John Wiley & Sons Ltd. The retraction has been made due to concerns relating to the validity of the EMG methodology and its interpretation, which affect the results of the paper. Reference Ferreira B, Da Silva GP, Gonçalves CR, Arnoni VW, Siéssere S, Semprini M, Verri ED, Chaves TC, Regalo SH. The effects of Duchenne muscular dystrophy on the performance of the stomatognathic system: case-control study. Developmental Medicine & Child Neurology 2015. doi:10.1111/dmcn.12707.
Collapse
|
38
|
Oxidative Stress-Mediated Skeletal Muscle Degeneration: Molecules, Mechanisms, and Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6842568. [PMID: 26798425 PMCID: PMC4700198 DOI: 10.1155/2016/6842568] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/08/2015] [Accepted: 10/08/2015] [Indexed: 11/25/2022]
Abstract
Oxidative stress is a loss of balance between the production of reactive oxygen species during cellular metabolism and the mechanisms that clear these species to maintain cellular redox homeostasis. Increased oxidative stress has been associated with muscular dystrophy, and many studies have proposed mechanisms that bridge these two pathological conditions at the molecular level. In this review, the evidence indicating a causal role of oxidative stress in the pathogenesis of various muscular dystrophies is revisited. In particular, the mediation of cellular redox status in dystrophic muscle by NF-κB pathway, autophagy, telomere shortening, and epigenetic regulation are discussed. Lastly, the current stance of targeting these pathways using antioxidant therapies in preclinical and clinical trials is examined.
Collapse
|
39
|
De Andrade PBM, Neff LA, Strosova MK, Arsenijevic D, Patthey-Vuadens O, Scapozza L, Montani JP, Ruegg UT, Dulloo AG, Dorchies OM. Caloric restriction induces energy-sparing alterations in skeletal muscle contraction, fiber composition and local thyroid hormone metabolism that persist during catch-up fat upon refeeding. Front Physiol 2015; 6:254. [PMID: 26441673 PMCID: PMC4584973 DOI: 10.3389/fphys.2015.00254] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 08/28/2015] [Indexed: 11/18/2022] Open
Abstract
Weight regain after caloric restriction results in accelerated fat storage in adipose tissue. This catch-up fat phenomenon is postulated to result partly from suppressed skeletal muscle thermogenesis, but the underlying mechanisms are elusive. We investigated whether the reduced rate of skeletal muscle contraction-relaxation cycle that occurs after caloric restriction persists during weight recovery and could contribute to catch-up fat. Using a rat model of semistarvation-refeeding, in which fat recovery is driven by suppressed thermogenesis, we show that contraction and relaxation of leg muscles are slower after both semistarvation and refeeding. These effects are associated with (i) higher expression of muscle deiodinase type 3 (DIO3), which inactivates tri-iodothyronine (T3), and lower expression of T3-activating enzyme, deiodinase type 2 (DIO2), (ii) slower net formation of T3 from its T4 precursor in muscles, and (iii) accumulation of slow fibers at the expense of fast fibers. These semistarvation-induced changes persisted during recovery and correlated with impaired expression of transcription factors involved in slow-twitch muscle development. We conclude that diminished muscle thermogenesis following caloric restriction results from reduced muscle T3 levels, alteration in muscle-specific transcription factors, and fast-to-slow fiber shift causing slower contractility. These energy-sparing effects persist during weight recovery and contribute to catch-up fat.
Collapse
Affiliation(s)
- Paula B M De Andrade
- Department of Medicine, Physiology, University of Fribourg Fribourg, Switzerland
| | - Laurence A Neff
- Pharmaceutical Biochemistry, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| | - Miriam K Strosova
- Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| | - Denis Arsenijevic
- Department of Medicine, Physiology, University of Fribourg Fribourg, Switzerland
| | - Ophélie Patthey-Vuadens
- Pharmaceutical Biochemistry, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland ; Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| | - Jean-Pierre Montani
- Department of Medicine, Physiology, University of Fribourg Fribourg, Switzerland
| | - Urs T Ruegg
- Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| | - Abdul G Dulloo
- Department of Medicine, Physiology, University of Fribourg Fribourg, Switzerland
| | - Olivier M Dorchies
- Pharmaceutical Biochemistry, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland ; Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, University of Lausanne Geneva, Switzerland
| |
Collapse
|
40
|
Passerieux E, Hayot M, Jaussent A, Carnac G, Gouzi F, Pillard F, Picot MC, Böcker K, Hugon G, Pincemail J, Defraigne JO, Verrips T, Mercier J, Laoudj-Chenivesse D. Effects of vitamin C, vitamin E, zinc gluconate, and selenomethionine supplementation on muscle function and oxidative stress biomarkers in patients with facioscapulohumeral dystrophy: a double-blind randomized controlled clinical trial. Free Radic Biol Med 2015; 81:158-69. [PMID: 25246239 DOI: 10.1016/j.freeradbiomed.2014.09.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/11/2014] [Accepted: 09/11/2014] [Indexed: 12/29/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disease characterized by progressive weakness and atrophy of specific skeletal muscles. As growing evidence suggests that oxidative stress may contribute to FSHD pathology, antioxidants that might modulate or delay oxidative insults could help in maintaining FSHD muscle function. Our primary objective was to test whether oral administration of vitamin C, vitamin E, zinc gluconate, and selenomethionine could improve the physical performance of patients with FSHD. Adult patients with FSHD (n=53) were enrolled at Montpellier University Hospital (France) in a randomized, double-blind, placebo-controlled pilot clinical trial. Patients were randomly assigned to receive 500 mg vitamin C, 400mg vitamin E, 25mg zinc gluconate and 200 μg selenomethionine (n=26), or matching placebo (n=27) once a day for 17 weeks. Primary outcomes were changes in the two-minute walking test (2-MWT), maximal voluntary contraction, and endurance limit time of the dominant and nondominant quadriceps (MVCQD, MVCQND, TlimQD, and TlimQND, respectively) after 17 weeks of treatment. Secondary outcomes were changes in the antioxidant status and oxidative stress markers. Although 2-MWT, MVCQ, and TlimQ were all significantly improved in the supplemented group at the end of the treatment compared to baseline, only MVCQ and TlimQ variations were significantly different between groups (MVCQD: P=0.011; MVCQND: P=0.004; TlimQD: P=0.028; TlimQND: P=0.011). Similarly, the vitamin C (P<0.001), vitamin E as α-tocopherol (P<0.001), vitamin C/vitamin E ratio (P=0.017), vitamin E γ/α ratio (P=0.022) and lipid peroxides (P<0.001) variations were significantly different between groups. In conclusion, vitamin E, vitamin C, zinc, and selenium supplementation has no significant effect on the 2-MWT, but improves MVCQ and TlimQ of both quadriceps by enhancing the antioxidant defenses and reducing oxidative stress. This trial was registered at clinicaltrials.gov (number: NCT01596803).
Collapse
Affiliation(s)
- Emilie Passerieux
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Maurice Hayot
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Audrey Jaussent
- Department of Biostatistics and Epidemiology, University Hospital of Montpellier, Montpellier, France
| | - Gilles Carnac
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Fares Gouzi
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Fabien Pillard
- Department of Respiratory Exploration and Department of Sports Medicine, Larrey University Hospital, Toulouse CEDEX, France
| | - Marie-Christine Picot
- Department of Biostatistics and Epidemiology, University Hospital, Montpellier, France and CIC 1001-INSERM
| | - Koen Böcker
- Alan Turing Institute Almere, The Netherlands
| | - Gerald Hugon
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Joel Pincemail
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Jean O Defraigne
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Theo Verrips
- Utrecht University, Department of Biology, The Netherlands
| | - Jacques Mercier
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Dalila Laoudj-Chenivesse
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France.
| |
Collapse
|
41
|
Buckner JL, Bowden SA, Mahan JD. Optimizing Bone Health in Duchenne Muscular Dystrophy. Int J Endocrinol 2015; 2015:928385. [PMID: 26124831 PMCID: PMC4466394 DOI: 10.1155/2015/928385] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 01/30/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder characterized by progressive muscle weakness, with eventual loss of ambulation and premature death. The approved therapy with corticosteroids improves muscle strength, prolongs ambulation, and maintains pulmonary function. However, the osteoporotic impact of chronic corticosteroid use further impairs the underlying reduced bone mass seen in DMD, leading to increased fragility fractures of long bones and vertebrae. These serious sequelae adversely affect quality of life and can impact survival. The current clinical issues relating to bone health and bone health screening methods in DMD are presented in this review. Diagnostic studies, including biochemical markers of bone turnover and bone mineral density by dual energy X-ray absorptiometry (DXA), as well as spinal imaging using densitometric lateral spinal imaging, and treatment to optimize bone health in patients with DMD are discussed. Treatment with bisphosphonates offers a method to increase bone mass in these children; oral and intravenous bisphosphonates have been used successfully although treatment is typically reserved for children with fractures and/or bone pain with low bone mass by DXA.
Collapse
Affiliation(s)
- Jason L. Buckner
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- *Jason L. Buckner:
| | - Sasigarn A. Bowden
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - John D. Mahan
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| |
Collapse
|
42
|
Diapocynin, a dimer of the NADPH oxidase inhibitor apocynin, reduces ROS production and prevents force loss in eccentrically contracting dystrophic muscle. PLoS One 2014; 9:e110708. [PMID: 25329652 PMCID: PMC4201587 DOI: 10.1371/journal.pone.0110708] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/24/2014] [Indexed: 11/19/2022] Open
Abstract
Elevation of intracellular Ca2+, excessive ROS production and increased phospholipase A2 activity contribute to the pathology in dystrophin-deficient muscle. Moreover, Ca2+, ROS and phospholipase A2, in particular iPLA2, are thought to potentiate each other in positive feedback loops. NADPH oxidases (NOX) have been considered as a major source of ROS in muscle and have been reported to be overexpressed in muscles of mdx mice. We report here on our investigations regarding the effect of diapocynin, a dimer of the commonly used NOX inhibitor apocynin, on the activity of iPLA2, Ca2+ handling and ROS generation in dystrophic myotubes. We also examined the effects of diapocynin on force production and recovery ability of isolated EDL muscles exposed to eccentric contractions in vitro, a damaging procedure to which dystrophic muscle is extremely sensitive. In dystrophic myotubes, diapocynin inhibited ROS production, abolished iPLA2 activity and reduced Ca2+ influx through stretch-activated and store-operated channels, two major pathways responsible for excessive Ca2+ entry in dystrophic muscle. Diapocynin also prevented force loss induced by eccentric contractions of mdx muscle close to the value of wild-type muscle and reduced membrane damage as seen by Procion orange dye uptake. These findings support the central role played by NOX-ROS in the pathogenic cascade leading to muscular dystrophy and suggest diapocynin as an effective NOX inhibitor that might be helpful for future therapeutic approaches.
Collapse
|
43
|
|
44
|
Urso ML. Anti-inflammatory interventions and skeletal muscle injury: benefit or detriment? J Appl Physiol (1985) 2013; 115:920-8. [DOI: 10.1152/japplphysiol.00036.2013] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exercise, eccentric contractions, acute trauma, and disease are all causal mechanisms of skeletal muscle injury. After skeletal muscle is injured, it undergoes sequential phases of degeneration, inflammation, regeneration, and fibrosis. Events that occur in response to inflammation trigger regenerative processes. However, since inflammation causes pain, decreases skeletal muscle function, has a negative effect on performance, and contributes to fibrosis, which is one of the leading causes of delayed regeneration, the general practice has been to reduce inflammation. The problem with this approach is that preventing inflammation may hinder recovery. Current treatment options for inflammation are not necessarily effective and, in some cases, they may be unsafe. This review focuses on the question of whether the most beneficial course of treatment should be to block inflammation or if it is sensible to allow inflammatory processes to progress naturally. If blocking inflammation is perceived as a beneficial approach, it is not yet known at what time point during the inflammatory response it is most sensible to interfere. To address these issues, this review evaluates the effects of various anti-inflammatory agents on recovery processes in response to exercise-induced, traumatic, and disease-associated models of skeletal muscle injury. A collective analysis such as this should lay the foundation for future work that systematically manipulates the inflammatory response to most effectively promote regeneration and functional recovery in injured skeletal muscle, while reducing the negative effects of inflammatory processes such as pain and fibrosis.
Collapse
Affiliation(s)
- Maria L. Urso
- United States Army Research Institute of Environmental Medicine, Military Performance Division, Natick, Massachusetts
| |
Collapse
|
45
|
Dorchies OM, Reutenauer-Patte J, Dahmane E, Ismail HM, Petermann O, Patthey- Vuadens O, Comyn SA, Gayi E, Piacenza T, Handa RJ, Décosterd LA, Ruegg UT. The anticancer drug tamoxifen counteracts the pathology in a mouse model of duchenne muscular dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:485-504. [PMID: 23332367 DOI: 10.1016/j.ajpath.2012.10.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 09/17/2012] [Accepted: 10/07/2012] [Indexed: 12/18/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a severe disorder characterized by progressive muscle wasting,respiratory and cardiac impairments, and premature death. No treatment exists so far, and the identification of active substances to fight DMD is urgently needed. We found that tamoxifen, a drug used to treat estrogen-dependent breast cancer, caused remarkable improvements of muscle force and of diaphragm and cardiac structure in the mdx(5Cv) mouse model of DMD. Oral tamoxifen treatment from 3 weeks of age for 15 months at a dose of 10 mg/kg/day stabilized myofiber membranes, normalized whole body force, and increased force production and resistance to repeated contractions of the triceps muscle above normal values. Tamoxifen improved the structure of leg muscles and diminished cardiac fibrosis by~ 50%. Tamoxifen also reduced fibrosis in the diaphragm, while increasing its thickness,myofiber count, and myofiber diameter, thereby augmenting by 72% the amount of contractile tissue available for respiratory function. Tamoxifen conferred a markedly slower phenotype to the muscles.Tamoxifen and its metabolites were present in nanomolar concentrations in plasma and muscles,suggesting signaling through high-affinity targets. Interestingly, the estrogen receptors ERa and ERb were several times more abundant in dystrophic than in normal muscles, and tamoxifen normalized the relative abundance of ERb isoforms. Our findings suggest that tamoxifen might be a useful therapy for DMD.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Behavior, Animal/drug effects
- Biomarkers/metabolism
- Biomechanical Phenomena/drug effects
- Body Weight/drug effects
- Creatine Kinase/blood
- Diaphragm/pathology
- Diaphragm/physiopathology
- Disease Models, Animal
- Feeding Behavior/drug effects
- Fibrosis
- Mice
- Muscle Contraction/drug effects
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Muscular Dystrophy, Animal/blood
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/physiopathology
- Muscular Dystrophy, Duchenne/blood
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Myocardium/pathology
- Organ Size/drug effects
- Receptors, Estrogen/metabolism
- Tamoxifen/blood
- Tamoxifen/pharmacology
- Tamoxifen/therapeutic use
Collapse
Affiliation(s)
- Olivier M Dorchies
- Department of Pharmacology, University of Geneva and University of Lausanne, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Park JH, Chung EJ, Kwon HJ, Im SS, Lim JG, Song DK. Protective effect of melatonin on TNF-α-induced muscle atrophy in L6 myotubes. J Pineal Res 2013; 54:417-425. [PMID: 23278522 DOI: 10.1111/jpi.12036] [Citation(s) in RCA: 12] [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: 10/16/2012] [Accepted: 12/03/2012] [Indexed: 01/22/2023]
Abstract
Muscle atrophy, characterized by decreased cell number and size, is a serious concern for patients afflicted with inflammatory diseases. Mounting evidence indicates that tumor necrosis factor alpha (TNF-α) plays a critical role in muscle atrophy in a number of clinical settings. We hypothesize that reactive oxygen species (ROS) mediate TNF-α-induced muscle cell death and hypotrophy. Recently, melatonin has attracted attention because of its free-radical scavenging and antioxidant properties. The aim of the current study was to evaluate the possible protective role of melatonin in TNF-α-induced muscle cell death and hypotrophy in rat L6 myotubes. To examine this possible role, L6 myotubes were exposed to various concentrations of recombinant TNF-α for 24 hr. We found that TNF-α at a concentration of 100 ng/mL induced ROS generation and decreased cell viability. Further analysis revealed that apoptosis, but not autophagy, may be important for TNF-α-induced cell death. Melatonin significantly attenuated TNF-α-induced ROS generation and apoptosis. In addition, decreased muscle fiber diameter and increased muscle cell proteolysis by TNF-α was highly attenuated by treatment with melatonin. The effects of melatonin were mediated neither through its plasmalemmal receptors nor by modulating the nuclear factor kappa B pathway activated by TNF-α. Taken together, these results suggest that TNF-α may mediate ROS-induced muscle cell death and hypotrophy and that melatonin may be a useful tool for protecting against muscle atrophy stemming from inflammatory diseases.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | | | | | | | | | | |
Collapse
|
47
|
Malik V, Rodino-Klapac LR, Mendell JR. Emerging drugs for Duchenne muscular dystrophy. Expert Opin Emerg Drugs 2012; 17:261-77. [PMID: 22632414 DOI: 10.1517/14728214.2012.691965] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is the most common, severe childhood form of muscular dystrophy. Treatment is limited to glucocorticoids that have the benefit of prolonging ambulation by approximately 2 years and preventing scoliosis. Finding a more satisfactory treatment should focus on maintaining long-term efficacy with a minimal side effect profile. AREAS COVERED Authors discuss different therapeutic strategies that have been used in pre-clinical and clinical settings. EXPERT OPINION Multiple treatment approaches have emerged. Most attractive are molecular-based therapies that can express the missing dystrophin protein (exon skipping or mutation suppression) or a surrogate gene product (utrophin). Other approaches include increasing the strength of muscles (myostatin inhibitors), reducing muscle fibrosis and decreasing oxidative stress. Additional targets include inhibiting NF-κB to reduce inflammation or promoting skeletal muscle blood flow and muscle contractility using phosphodiesterase inhibitors or nitric oxide (NO) donors. The potential for each of these treatment strategies to enter clinical trials is a central theme of discussion. The review emphasizes that the goal of treatment should be to find a product at least as good as glucocorticoids with a lower side effect profile or with a significant glucocorticoid sparing effect.
Collapse
Affiliation(s)
- Vinod Malik
- The Ohio State University, Research Institute, Nationwide Children's Hospital and, Department of Pediatrics, Columbus, OH 43205, USA
| | | | | |
Collapse
|
48
|
Perkins KJ, Davies KE. Recent advances in Duchenne muscular dystrophy. Degener Neurol Neuromuscul Dis 2012; 2:141-164. [PMID: 30890885 DOI: 10.2147/dnnd.s26637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an allelic X-linked progressive muscle-wasting disease, is one of the most common single-gene disorders in the developed world. Despite knowledge of the underlying genetic causation and resultant pathophysiology from lack of dystrophin protein at the muscle sarcolemma, clinical intervention is currently restricted to symptom management. In recent years, however, unprecedented advances in strategies devised to correct the primary defect through gene- and cell-based therapeutics hold particular promise for treating dystrophic muscle. Conventional gene replacement and endogenous modification strategies have greatly benefited from continued improvements in encapsidation capacity, transduction efficiency, and systemic delivery. In particular, RNA-based modifying approaches such as exon skipping enable expression of a shorter but functional dystrophin protein and rapid progress toward clinical application. Emerging combined gene- and cell-therapy strategies also illustrate particular promise in enabling ex vivo genetic correction and autologous transplantation to circumvent a number of immune challenges. These approaches are complemented by a vast array of pharmacological approaches, in particular the successful identification of molecules that enable functional replacement or ameliorate secondary DMD pathology. Animal models have been instrumental in providing proof of principle for many of these strategies, leading to several recent trials that have investigated their efficacy in DMD patients. Although none has reached the point of clinical use, rapid improvements in experimental technology and design draw this goal ever closer. Here, we review therapeutic approaches to DMD, with particular emphasis on recent progress in strategic development, preclinical evaluation and establishment of clinical efficacy. Further, we discuss the numerous challenges faced and synergistic approaches being devised to combat dystrophic pathology effectively.
Collapse
Affiliation(s)
- Kelly J Perkins
- Sir William Dunn School of Pathology.,MRC Functional Genomics Unit, University of Oxford, Oxford, UK,
| | - Kay E Davies
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK,
| |
Collapse
|
49
|
Nakae Y, Dorchies OM, Stoward PJ, Zimmermann BF, Ritter C, Ruegg UT. Quantitative evaluation of the beneficial effects in the mdx mouse of epigallocatechin gallate, an antioxidant polyphenol from green tea. Histochem Cell Biol 2012; 137:811-27. [PMID: 22331205 PMCID: PMC3353109 DOI: 10.1007/s00418-012-0926-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2012] [Indexed: 12/17/2022]
Abstract
In two separate previous studies, we reported that subcutaneous (sc) or oral administration of (−)-epigallocatechin-3-gallate (EGCG) limited the development of muscle degeneration of mdx mice, a mild phenotype model for Duchenne muscular dystrophy (DMD). However, it was not possible to conclude which was the more efficient route of EGCG administration because different strains of mdx mice, periods of treatment and methods of assessment were used. In this study, we investigated which administration routes and dosages of EGCG are the most effective for limiting the onset of dystrophic lesions in the same strain of mdx mice and applying the same methods of assessment. Three-week-old mdx mice were injected sc for 5 weeks with either saline or a daily average of 3 or 6 mg/kg EGCG. For comparison, age-matched mdx mice were fed for 5 weeks with either a diet containing 0.1% EGCG or a control diet. The effects of EGCG were assessed quantitatively by determining the activities of serum muscle-derived creatine kinase, isometric contractions of triceps surae muscles, integrated spontaneous locomotor activities, and oxidative stress and fibrosis in selected muscles. Oral administration of 180 mg/kg/day EGCG in the diet was found the most effective for significantly improving several parameters associated with muscular dystrophy. However, the improvements were slightly less than those observed previously for sc injection started immediately after birth. The efficacy of EGCG for limiting the development of dystrophic muscle lesions in mice suggests that EGCG may be of benefit for DMD patients.
Collapse
Affiliation(s)
- Yoshiko Nakae
- Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, 30 Quai Ernest Ansermet, 1211, Geneva 4, Switzerland.
| | | | | | | | | | | |
Collapse
|