1
|
Li Z, Wang Y, Huang W, Shi X, Ma T, Yu X. miR-155 induces sepsis-associated damage to the intestinal mucosal barrier via sirtuin 1/nuclear factor-κB-mediated intestinal pyroptosis. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39262326 DOI: 10.3724/abbs.2024124] [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: 09/13/2024] Open
Abstract
Sepsis is a life-threatening state of organ dysfunction caused by systemic inflammation and a dysfunctional response to host infections that can induce severe intestinal mucosal damage. Pyroptosis is mediated by the activated NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome after stimulation by various inflammatory factors during sepsis. The inflammatory response is a major driver of intestinal damage during sepsis. Intestinal mucosal barrier dysfunction in sepsis is associated with pyroptosis, a type of programmed inflammatory cell death. Several studies have confirmed the role of miR-155 in sepsis and other diseases. However, the effect of miR-155 on intestinal pyroptosis in the context of intestinal mucosal barrier dysfunction during sepsis remains unclear. Thus, a model of sepsis in Sprague-Dawley rats is established using cecal ligation and puncture (CLP), and a series of molecular biological methods are used in this study. The results show that the expression of miR-155 is increased and that of sirtuin 1 (SIRT1) is decreased in the intestinal tissues of patients with sepsis. miR-155 expression is negatively correlated with SIRT1 expression. Increased miR-155 expression significantly inhibits SIRT1 activity and upregulates the expressions of NOD-like receptor family pyrin domain-containing 3 (NLRP3), caspase-1, apoptosis-associated speck-like protein containing a CARD (ASC), interleukin-1β (IL-1β) and interleukin-18 (IL-18) to promote pyroptosis. The inhibition of miR-155 expression is associated with increased SIRT1 expression, promotes the deacetylation of p65, and significantly downregulates p65 acetylation. Herein, we propose that miR-155 induces pyroptosis in the intestine partly by regulating SIRT1, thereby reducing the deacetylation of the nuclear factor (NF)-κB subunit p65 and increasing NF-κB signaling activity in sepsis, leading to intestinal barrier damage.
Collapse
Affiliation(s)
- Zhihua Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Yi Wang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
- Department of Critical Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Weiwei Huang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Xingyu Shi
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Tao Ma
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Xiangyou Yu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
- Department of Critical Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
2
|
Jin X, Zhang Y, Zhou Y, Luo Y, Han X, Gao Y, Yu H, Duan Y, Shi L, Wu Y, Li Y. Sirt1 Deficiency Promotes Age-Related AF Through Enhancing Atrial Necroptosis by Activation of RIPK1 Acetylation. Circ Arrhythm Electrophysiol 2024; 17:e012452. [PMID: 39012929 DOI: 10.1161/circep.123.012452] [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: 09/06/2023] [Accepted: 05/16/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Aging is one of the most potent risk determinants for the onset of atrial fibrillation (AF). Sirts (sirtuins) have been implicated in the pathogenesis of cardiovascular disease, and their expression declines with aging. However, whether Sirts involved in age-related AF and its underlying mechanisms remain unknown. The present study aims to explore the role of Sirts in age-related AF and delineate the underlying molecular mechanisms. METHODS Sirt1 levels in the atria of both elderly individuals and aging rats were evaluated using quantitative real-time polymerase chain reaction and Western blot analysis. Mice were engineered to specifically knockout Sirt1 in the atria and right ventricle (Sirt1mef2c/mef2c). Various techniques, such as echocardiography, atrial electrophysiology, and protein acetylation modification omics were employed. Additionally, coimmunoprecipitation was utilized to substantiate the interaction between Sirt1 and RIPK1 (receptor-interacting protein kinase 1). RESULTS We discerned that among the diverse subtypes of sirtuin proteins, only Sirt1 expression was significantly diminished in the atria of elderly people and aged rats. The Sirt1mef2c/mef2c mice exhibited an enlarged atrial diameter and heightened vulnerability to AF. Acetylated proteomics and cell experiments identified that Sirt1 deficiency activated atrial necroptosis through increasing RIPK1 acetylation and subsequent pseudokinase MLKL (mixed lineage kinase domain-like protein) phosphorylation. Consistently, necroptotic inhibitor necrosulfonamide mitigated atrial necroptosis and diminished both the atrial diameter and AF susceptibility of Sirt1mef2c/mef2c mice. Resveratrol prevented age-related AF in rats by activating atrial Sirt1 and inhibiting necroptosis. CONCLUSIONS Our findings first demonstrated that Sirt1 exerts significant efficacy in countering age-related AF by impeding atrial necroptosis through regulation of RIPK1 acetylation, highlighting that the activation of Sirt1 or the inhibition of necroptosis could potentially serve as a therapeutic strategy for age-related AF.
Collapse
Affiliation(s)
- Xuexin Jin
- Department of Cardiology the First Affiliated Hospital of Harbin Medical University (X.J., Y. Zhang, Y. Zhou, Y. Li)
| | - Yun Zhang
- Department of Cardiology the First Affiliated Hospital of Harbin Medical University (X.J., Y. Zhang, Y. Zhou, Y. Li)
| | - Yun Zhou
- Department of Cardiology the First Affiliated Hospital of Harbin Medical University (X.J., Y. Zhang, Y. Zhou, Y. Li)
| | - Yingchun Luo
- NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital of Harbin Medical University (Y. Luo, X.H., Y.G.)
| | - Xuejie Han
- NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital of Harbin Medical University (Y. Luo, X.H., Y.G.)
| | - Yunlong Gao
- NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital of Harbin Medical University (Y. Luo, X.H., Y.G.)
| | - Hui Yu
- Key Laboratory of Cardiac Diseases & Heart Failure (H.Y., Y.D., L.S.)
| | - Yu Duan
- Key Laboratory of Cardiac Diseases & Heart Failure (H.Y., Y.D., L.S.)
| | - Ling Shi
- Key Laboratory of Cardiac Diseases & Heart Failure (H.Y., Y.D., L.S.)
| | - Yue Wu
- Department of Cardiology, the First Hospital of Xi'an Jiaotong University, Xi'an, China (Y.W.)
| | - Yue Li
- Department of Cardiology the First Affiliated Hospital of Harbin Medical University (X.J., Y. Zhang, Y. Zhou, Y. Li)
- State Key Laboratory of Frigid Zone Cardiovascular Disease (Y. Li), Harbin Medical University
- Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases (Y. Li)
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin (Y. Li)
| |
Collapse
|
3
|
Casati SR, Cervia D, Roux-Biejat P, Moscheni C, Perrotta C, De Palma C. Mitochondria and Reactive Oxygen Species: The Therapeutic Balance of Powers for Duchenne Muscular Dystrophy. Cells 2024; 13:574. [PMID: 38607013 PMCID: PMC11011272 DOI: 10.3390/cells13070574] [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: 02/19/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic progressive muscle-wasting disorder that leads to rapid loss of mobility and premature death. The absence of functional dystrophin in DMD patients reduces sarcolemma stiffness and increases contraction damage, triggering a cascade of events leading to muscle cell degeneration, chronic inflammation, and deposition of fibrotic and adipose tissue. Efforts in the last decade have led to the clinical approval of novel drugs for DMD that aim to restore dystrophin function. However, combination therapies able to restore dystrophin expression and target the myriad of cellular events found impaired in dystrophic muscle are desirable. Muscles are higher energy consumers susceptible to mitochondrial defects. Mitochondria generate a significant source of reactive oxygen species (ROS), and they are, in turn, sensitive to proper redox balance. In both DMD patients and animal models there is compelling evidence that mitochondrial impairments have a key role in the failure of energy homeostasis. Here, we highlighted the main aspects of mitochondrial dysfunction and oxidative stress in DMD and discussed the recent findings linked to mitochondria/ROS-targeted molecules as a therapeutic approach. In this respect, dual targeting of both mitochondria and redox homeostasis emerges as a potential clinical option in DMD.
Collapse
Affiliation(s)
- Silvia Rosanna Casati
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via Fratelli Cervi 93, 20054 Segrate, Italy; (S.R.C.); (C.D.P.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; (P.R.-B.); (C.M.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; (P.R.-B.); (C.M.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; (P.R.-B.); (C.M.)
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via Fratelli Cervi 93, 20054 Segrate, Italy; (S.R.C.); (C.D.P.)
| |
Collapse
|
4
|
Nakashima R, Hosoda R, Tatekoshi Y, Iwahara N, Saga Y, Kuno A. Transcriptional dysregulation of autophagy in the muscle of a mouse model of Duchenne muscular dystrophy. Sci Rep 2024; 14:1365. [PMID: 38228650 DOI: 10.1038/s41598-024-51746-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
It has been reported that autophagic activity is disturbed in the skeletal muscles of dystrophin-deficient mdx mice and patients with Duchenne muscular dystrophy (DMD). Transcriptional regulations of autophagy by FoxO transcription factors (FoxOs) and transcription factor EB (TFEB) play critical roles in adaptation to cellular stress conditions. Here, we investigated whether autophagic activity is dysregulated at the transcription level in dystrophin-deficient muscles. Expression levels of autophagy-related genes were globally decreased in tibialis anterior and soleus muscles of mdx mice compared with those of wild-type mice. DNA microarray data from the NCBI database also showed that genes related to autophagy were globally downregulated in muscles from patients with DMD. These downregulated genes are known as targets of FoxOs and TFEB. Immunostaining showed that nuclear localization of FoxO1 and FoxO3a was decreased in mdx mice. Western blot analyses demonstrated increases in phosphorylation levels of FoxO1 and FoxO3a in mdx mice. Nuclear localization of TFEB was also reduced in mdx mice, which was associated with elevated phosphorylation levels of TFEB. Collectively, the results suggest that autophagy is disturbed in dystrophin-deficient muscles via transcriptional downregulation due to phosphorylation-mediated suppression of FoxOs and TFEB.
Collapse
Affiliation(s)
- Ryuta Nakashima
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Yuki Tatekoshi
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
- Department of Neurology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yukika Saga
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
5
|
Cree T, Gomez TR, Timpani CA, Rybalka E, Price JT, Goodman CA. FKBP25 regulates myoblast viability and migration and is differentially expressed in in vivo models of muscle adaptation. FEBS J 2023; 290:4660-4678. [PMID: 37345229 DOI: 10.1111/febs.16894] [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: 09/01/2022] [Revised: 04/18/2023] [Accepted: 06/21/2023] [Indexed: 06/23/2023]
Abstract
FKBP25 (FKBP3 gene) is a dual-domain PPIase protein that consists of a C-terminal PPIase domain and an N-terminal basic tilted helix bundle (BTHB). The PPIase domain of FKBP25 has been shown to bind to microtubules, which has impacts upon microtubule polymerisation and cell cycle progression. Using quantitative proteomics, it was recently found that FKBP25 was expressed in the top 10% of the mouse skeletal muscle proteome. However, to date there have been few studies investigating the role of FKBP25 in non-transformed systems. As such, this study aimed to investigate potential roles for FKBP25 in myoblast viability, migration and differentiation and in adaptation of mature skeletal muscle. Doxycycline-inducible FKBP25 knockdown in C2C12 myoblasts revealed an increase in cell accumulation/viability and migration in vitro that was independent of alterations in tubulin dynamics; however, FKBP25 knockdown had no discernible impact on myoblast differentiation into myotubes. Finally, a series of in vivo models of muscle adaptation were assessed, where it was observed that FKBP25 protein expression was increased in hypertrophy and regeneration conditions (chronic mechanical overload and the mdx model of Duchenne muscular dystrophy) but decreased in an atrophy model (denervation). Overall, the findings of this study establish FKBP25 as a regulator of myoblast viability and migration, with possible implications for satellite cell proliferation and migration and muscle regeneration, and as a potential regulator of in vivo skeletal muscle adaptation.
Collapse
Affiliation(s)
- Tabitha Cree
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - Tania Ruz Gomez
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
| | - Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - John T Price
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
- Monash Biomedicine Discovery Institute, Clayton, Australia
| | - Craig A Goodman
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Parkville, Australia
| |
Collapse
|
6
|
Drummond SE, Burns DP, El Maghrani S, Ziegler O, Healy V, O'Halloran KD. Chronic Intermittent Hypoxia-Induced Diaphragm Muscle Weakness Is NADPH Oxidase-2 Dependent. Cells 2023; 12:1834. [PMID: 37508499 PMCID: PMC10377874 DOI: 10.3390/cells12141834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/21/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic intermittent hypoxia (CIH)-induced redox alterations underlie diaphragm muscle dysfunction. We sought to establish if NADPH oxidase 2 (NOX2)-derived reactive oxygen species (ROS) underpin CIH-induced changes in diaphragm muscle, which manifest as impaired muscle performance. Adult male mice (C57BL/6J) were assigned to one of three groups: normoxic controls (sham); chronic intermittent hypoxia-exposed (CIH, 12 cycles/hour, 8 h/day for 14 days); and CIH + apocynin (NOX2 inhibitor, 2 mM) administered in the drinking water throughout exposure to CIH. In separate studies, we examined sham and CIH-exposed NOX2-null mice (B6.129S-CybbTM1Din/J). Apocynin co-treatment or NOX2 deletion proved efficacious in entirely preventing diaphragm muscle dysfunction following exposure to CIH. Exposure to CIH had no effect on NOX2 expression. However, NOX4 mRNA expression was increased following exposure to CIH in wild-type and NOX2 null mice. There was no evidence of overt CIH-induced oxidative stress. A NOX2-dependent increase in genes related to muscle regeneration, antioxidant capacity, and autophagy and atrophy was evident following exposure to CIH. We suggest that NOX-dependent CIH-induced diaphragm muscle weakness has the potential to affect ventilatory and non-ventilatory performance of the respiratory system. Therapeutic strategies employing NOX2 blockade may function as an adjunct therapy to improve diaphragm muscle performance and reduce disease burden in diseases characterised by exposure to CIH, such as obstructive sleep apnoea.
Collapse
Affiliation(s)
- Sarah E Drummond
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - Sarah El Maghrani
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - Oscar Ziegler
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - Vincent Healy
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| |
Collapse
|
7
|
Xie K, Yang Q, Yan Z, Gao X, Huang X, Wang P, Li J, Li J, Wang Z, Gao Y, Gun S. Overexpression of SIRT1 alleviates oxidative damage and barrier dysfunction in CPB2 toxin-infected IPEC-J2 cells. Microb Pathog 2023:106181. [PMID: 37276895 DOI: 10.1016/j.micpath.2023.106181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023]
Abstract
Clostridium perfringens (C. perfringens) beta2 (CPB2) toxin may induce necrotizing enteritis (NE) in pigs. Sirtuin1 (SIRT1) is involved in inflammatory intestinal diseases and affects intestinal barrier function. However, the effects of SIRT1 on piglet intestinal disease caused by CPB2 toxin are unclear. This study revealed the role of pig SIRT1 in CPB2 toxin-exposed intestinal porcine epithelial cells (IPEC-J2). Herein, we manifested that SIRT1 was dramatically decreased in IPEC-J2 cells infected with CPB2 toxin. Subsequently, we silenced and overexpressed SIRT1 using siRNA and a overexpression vector in CPB2 toxin-treated IPEC-J2 cells. The results indicated that overexpression of SIRT1 suppressed reactive oxygen species (ROS) generates, the expression tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and Bax, nuclear factor-kappa B (NF-κB p65), phospho (p)-NF-kB p65 and lactate dehydrogenase (LDH) activity and apoptosis in CPB2 toxin-treated IPEC-J2 cells, and increased IL-10, mitochondrial membrane potential (ΔΨm), Bcl-2, Claudin1 and Occludin levels and cell viability. These results indicated that SIRT1 protects IPEC-J2 cells against CPB2 toxin-induced oxidative damage and tight junction (TJ) disruption, which provides a theoretical basis for further study of the molecular regulatory mechanism of SIRT1 in C. perfringens-infected NE in piglets.
Collapse
Affiliation(s)
- Kaihui Xie
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiyou Li
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, 730070, China
| | - Zike Wang
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, 730070, China
| | - Yi Gao
- Jilin Rongtai Agricultural Development Co, Ltd, Changchun, Jilin, 130507, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, 730070, China.
| |
Collapse
|
8
|
Hosoda R, Nakashima R, Yano M, Iwahara N, Asakura S, Nojima I, Saga Y, Kunimoto R, Horio Y, Kuno A. Resveratrol, a SIRT1 activator, attenuates aging-associated alterations in skeletal muscle and heart in mice. J Pharmacol Sci 2023; 152:112-122. [PMID: 37169475 DOI: 10.1016/j.jphs.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Aging is associated with impairment of multiple organs, including skeletal muscle and heart. In this study, we investigated whether resveratrol, an activator of an NAD+-dependent protein deacetylase Sirtuin-1 (SIRT1), attenuates age-related sarcopenia and cardiomyocyte hypertrophy in mice. Treatment of mice with resveratrol (0.4 g/kg diet) from 28 weeks of age for 32 weeks prevented aging-associated shortening of rotarod riding time. In the tibialis anterior (TA) muscle, histogram analysis showed that the atrophic muscle was increased in 60-week-old (wo) mice compared with 20-wo mice, which was attenuated by resveratrol. In the heart, resveratrol attenuated an aging-associated increase in the cardiomyocyte diameter. Acetylated proteins were increased and autophagic activity was reduced in the TA muscle of 60-wo mice compared with those of 20-wo mice. Resveratrol treatment reduced levels of acetylated proteins and restored autophagic activity in the TA muscle. Aging-related reduction in myocardial autophagy was also suppressed by resveratrol. Skeletal muscle-specific SIRT1 knockout mice showed increases in acetylated proteins and atrophic muscle fibers and reduced autophagic activity in the TA muscle. These results suggest that activation of SIRT1 by treatment with resveratrol suppresses sarcopenia and cardiomyocyte hypertrophy by restoration of autophagy in mice.
Collapse
Affiliation(s)
- Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Ryuta Nakashima
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Masaki Yano
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Seidai Asakura
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Iyori Nojima
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Yukika Saga
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Risa Kunimoto
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan.
| |
Collapse
|
9
|
Sandonà M, Cavioli G, Renzini A, Cedola A, Gigli G, Coletti D, McKinsey TA, Moresi V, Saccone V. Histone Deacetylases: Molecular Mechanisms and Therapeutic Implications for Muscular Dystrophies. Int J Mol Sci 2023; 24:4306. [PMID: 36901738 PMCID: PMC10002075 DOI: 10.3390/ijms24054306] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Histone deacetylases (HDACs) are enzymes that regulate the deacetylation of numerous histone and non-histone proteins, thereby affecting a wide range of cellular processes. Deregulation of HDAC expression or activity is often associated with several pathologies, suggesting potential for targeting these enzymes for therapeutic purposes. For example, HDAC expression and activity are higher in dystrophic skeletal muscles. General pharmacological blockade of HDACs, by means of pan-HDAC inhibitors (HDACi), ameliorates both muscle histological abnormalities and function in preclinical studies. A phase II clinical trial of the pan-HDACi givinostat revealed partial histological improvement and functional recovery of Duchenne Muscular Dystrophy (DMD) muscles; results of an ongoing phase III clinical trial that is assessing the long-term safety and efficacy of givinostat in DMD patients are pending. Here we review the current knowledge about the HDAC functions in distinct cell types in skeletal muscle, identified by genetic and -omic approaches. We describe the signaling events that are affected by HDACs and contribute to muscular dystrophy pathogenesis by altering muscle regeneration and/or repair processes. Reviewing recent insights into HDAC cellular functions in dystrophic muscles provides new perspectives for the development of more effective therapeutic approaches based on drugs that target these critical enzymes.
Collapse
Affiliation(s)
| | - Giorgia Cavioli
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessia Cedola
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), 73100 Lecce, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
- CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Aging B2A, Sorbonne Université, 75005 Paris, France
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Viviana Moresi
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Valentina Saccone
- IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
10
|
Chen XM, Liu YH, Ji SF, Xue XM, Wang LL, Zhang M, Chang YM, Wang XC. Protective effect of ginsenoside Rd on military aviation noise-induced cochlear hair cell damage in guinea pigs. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:23965-23981. [PMID: 36331733 DOI: 10.1007/s11356-022-23504-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Noise pollution has become one of the important social hazards that endanger the auditory system of residents, causing noise-induced hearing loss (NIHL). Oxidative stress has a significant role in the pathogenesis of NIHL, in which the silent information regulator 1(SIRT1)/proliferator-activated receptor-gamma coactivator 1α (PGC-1α) signaling pathway is closely engaged. Ginsenoside Rd (GSRd), a main monomer extract from ginseng plants, has been confirmed to suppress oxidative stress. Therefore, the hypothesis that GSRd may attenuate noise-induced cochlear hair cell loss seemed promising. Forty-eight male guinea pigs were randomly divided into four groups: control, noise exposure, GSRd treatment (30 mg/kg Rd for 10d + noise), and experimental control (30 mg/kg glycerol + noise). The experimental groups received military helicopter noise exposure at 115 dB (A) for 4 h daily for five consecutive days. Hair cell damage was evaluated by using inner ear basilar membrane preparation and scanning electron microscopy. Terminal dUTP nick end labeling (TUNEL) and immunofluorescence staining were conducted. Changes in the SIRT1/PGC-1α signaling pathway and other apoptosis-related markers in the cochleae, as well as oxidative stress parameters, were used as readouts. Loss of outer hair cells, more disordered cilia, prominent apoptosis, and elevated free radical levels were observed in the experimental groups. GSRd treatment markedly mitigated hearing threshold shifts, ameliorated outer hair cell loss and lodging or loss of cilia, and improved apoptosis through decreasing Bcl-2 associated X protein (Bax) expression and increasing Bcl-2 expression. In addition, GSRd alleviated the noise-induced cochlear redox injury by upregulating superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels, decreasing malondialdehyde (MDA) levels, and enhancing the activity of SIRT1 and PGC-1α messenger ribonucleic acid (mRNA) and protein expression. In conclusion, GSRd can improve structural and oxidative damage to the cochleae caused by noise. The underlying mechanisms may be associated with the SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Xue-Min Chen
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Yu-Hui Liu
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an, China
- Department of Aviation Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
- Air Force Health Care Center for Special Services, Hangzhou, China
| | - Shuai-Fei Ji
- Medical School of Chinese PLA, Beijing, China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xin-Miao Xue
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Lin-Lin Wang
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Min Zhang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an, China
- Department of Aviation Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Yao-Ming Chang
- Department of Aerospace Hygiene, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Xiao-Cheng Wang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an, China.
- Department of Aviation Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi'an, China.
| |
Collapse
|
11
|
Dubuisson N, Versele R, Planchon C, Selvais CM, Noel L, Abou-Samra M, Davis-López de Carrizosa MA. Histological Methods to Assess Skeletal Muscle Degeneration and Regeneration in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:16080. [PMID: 36555721 PMCID: PMC9786356 DOI: 10.3390/ijms232416080] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease caused by the loss of function of the protein dystrophin. This protein contributes to the stabilisation of striated cells during contraction, as it anchors the cytoskeleton with components of the extracellular matrix through the dystrophin-associated protein complex (DAPC). Moreover, absence of the functional protein affects the expression and function of proteins within the DAPC, leading to molecular events responsible for myofibre damage, muscle weakening, disability and, eventually, premature death. Presently, there is no cure for DMD, but different treatments help manage some of the symptoms. Advances in genetic and exon-skipping therapies are the most promising intervention, the safety and efficiency of which are tested in animal models. In addition to in vivo functional tests, ex vivo molecular evaluation aids assess to what extent the therapy has contributed to the regenerative process. In this regard, the later advances in microscopy and image acquisition systems and the current expansion of antibodies for immunohistological evaluation together with the development of different spectrum fluorescent dyes have made histology a crucial tool. Nevertheless, the complexity of the molecular events that take place in dystrophic muscles, together with the rise of a multitude of markers for each of the phases of the process, makes the histological assessment a challenging task. Therefore, here, we summarise and explain the rationale behind different histological techniques used in the literature to assess degeneration and regeneration in the field of dystrophinopathies, focusing especially on those related to DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Neuromuscular Reference Center, Cliniques Universitaires Saint-Luc (CUSL), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
12
|
The Beneficial Role of Sirtuin 1 in Preventive or Therapeutic Options of Neurodegenerative Diseases. Neuroscience 2022; 504:79-92. [DOI: 10.1016/j.neuroscience.2022.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/08/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
|
13
|
Activation of SIRT1 promotes membrane resealing via cortactin. Sci Rep 2022; 12:15328. [PMID: 36097021 PMCID: PMC9468153 DOI: 10.1038/s41598-022-19136-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Muscular dystrophies are inherited myopathic disorders characterized by progressive muscle weakness. Recently, several gene therapies have been developed; however, the treatment options are still limited. Resveratrol, an activator of SIRT1, ameliorates muscular function in muscular dystrophy patients and dystrophin-deficient mdx mice, although its mechanism is still not fully elucidated. Here, we investigated the effects of resveratrol on membrane resealing. We found that resveratrol promoted membrane repair in C2C12 cells via the activation of SIRT1. To elucidate the mechanism by which resveratrol promotes membrane resealing, we focused on the reorganization of the cytoskeleton, which occurs in the early phase of membrane repair. Treatment with resveratrol promoted actin accumulation at the injured site. We also examined the role of cortactin in membrane resealing. Cortactin accumulated at the injury site, and cortactin knockdown suppressed membrane resealing and reorganization of the cytoskeleton. Additionally, SIRT1 deacetylated cortactin and promoted the interaction between cortactin and F-actin, thus possibly enhancing the accumulation of cortactin at the injury site. Finally, we performed a membrane repair assay using single fiber myotubes from control and resveratrol-fed mice, where the oral treatment with resveratrol promoted membrane repair ex vivo. These findings suggest that resveratrol promotes membrane repair via the SIRT1/cortactin axis.
Collapse
|
14
|
Zhou S, Xue J, Shan J, Hong Y, Zhu W, Nie Z, Zhang Y, Ji N, Luo X, Zhang T, Ma W. Gut-Flora-Dependent Metabolite Trimethylamine-N-Oxide Promotes Atherosclerosis-Associated Inflammation Responses by Indirect ROS Stimulation and Signaling Involving AMPK and SIRT1. Nutrients 2022; 14:3338. [PMID: 36014845 PMCID: PMC9416570 DOI: 10.3390/nu14163338] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trimethylamine-N-oxide (TMAO), a gut-microbiota-dependent metabolite after ingesting dietary choline, has been identified as a novel risk factor for atherosclerosis through inducing vascular inflammation. However, the underlying molecular mechanism is poorly understood. Using an in vitro vascular cellular model, we found that the TMAO-induced inflammation responses were correlated with an elevation of ROS levels and downregulation of SIRT1 expression in VSMCs and HUVECs. The overexpression of SIRT1 could abrogate both the stimulation of ROS and inflammation. Further studies revealed that AMPK was also suppressed by TMAO and was a mediator upstream of SIRT1. Activation of AMPK by AICAR could reduce TMAO-induced ROS and inflammation. Moreover, the GSH precursor NAC could attenuate TMAO-induced inflammation. In vivo studies with mice models also showed that choline-induced production of TMAO and the associated glycolipid metabolic changes leading to atherosclerosis could be relieved by NAC and a probiotic LP8198. Collectively, the present study revealed an unrecognized mechanistic link between TMAO and atherosclerosis risk, and probiotics ameliorated TMAO-induced atherosclerosis through affecting the gut microbiota. Consistent with previous studies, our data confirmed that TMAO could stimulate inflammation by modulating cellular ROS levels. However, this was not due to direct cytotoxicity but through complex signaling pathways involving AMPK and SIRT1.
Collapse
Affiliation(s)
- Sa Zhou
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiamin Xue
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingbo Shan
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yingxiang Hong
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wenkang Zhu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhiyan Nie
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yujie Zhang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nanxi Ji
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongcun Zhang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Wenjian Ma
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Qilu Institute of Technology, Jinan 250200, China
| |
Collapse
|
15
|
Georgieva AM, Guo X, Bartkuhn M, Günther S, Künne C, Smolka C, Atzberger A, Gärtner U, Mamchaoui K, Bober E, Zhou Y, Yuan X, Braun T. Inactivation of Sirt6 ameliorates muscular dystrophy in mdx mice by releasing suppression of utrophin expression. Nat Commun 2022; 13:4184. [PMID: 35859073 PMCID: PMC9300598 DOI: 10.1038/s41467-022-31798-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
The NAD+-dependent SIRT1-7 family of protein deacetylases plays a vital role in various molecular pathways related to stress response, DNA repair, aging and metabolism. Increased activity of individual sirtuins often exerts beneficial effects in pathophysiological conditions whereas reduced activity is usually associated with disease conditions. Here, we demonstrate that SIRT6 deacetylates H3K56ac in myofibers to suppress expression of utrophin, a dystrophin-related protein stabilizing the sarcolemma in absence of dystrophin. Inactivation of Sirt6 in dystrophin-deficient mdx mice reduced damage of myofibers, ameliorated dystrophic muscle pathology, and improved muscle function, leading to attenuated activation of muscle stem cells (MuSCs). ChIP-seq and locus-specific recruitment of SIRT6 using a CRISPR-dCas9/gRNA approach revealed that SIRT6 is critical for removal of H3K56ac at the Downstream utrophin Enhancer (DUE), which is indispensable for utrophin expression. We conclude that epigenetic manipulation of utrophin expression is a promising approach for the treatment of Duchenne Muscular Dystrophy (DMD). Utrophin is a dystrophin-related protein stabilizing the sarcolemma in absence of dystrophin. Here the authors report that inactivation of the protein deacetylase SIRT6, involved in the deacetylation of the epigenetic mark H3K56ac in muscle cells, increases expression of utrophin and ameliorates dystrophic muscle pathology in mice.
Collapse
Affiliation(s)
- Angelina M Georgieva
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Xinyue Guo
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Justus Liebig University, Giessen, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Carsten Künne
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Christian Smolka
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Ann Atzberger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Ulrich Gärtner
- Institute for Anatomy and Cell Biology, University of Giessen, Giessen, Germany
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013, Paris, France
| | - Eva Bober
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Yonggang Zhou
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
| |
Collapse
|
16
|
Wang R, Yuan W, Li L, Lu F, Zhang L, Gong H, Huang X. Resveratrol ameliorates muscle atrophy in chronic kidney disease via the axis of SIRT1/FoxO1. Phytother Res 2022; 36:3265-3275. [PMID: 35606908 DOI: 10.1002/ptr.7499] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/28/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022]
Abstract
Chronic kidney disease (CKD) is often associated with muscle atrophy. However, the underlying molecular mechanisms are still not well understood. Here, we treated 5/6-nephrectomized (5/6Nx) rats with resveratrol and found that this treatment greatly improves renal function as evidenced by reduced proteinuria and cystatin C. Moreover, resveratrol ameliorates renal fibrosis by reducing transforming growth factor β (TGF-β) and connective tissue growth factor (CTGF). Meanwhile, muscle atrophy in these 5/6Nx rats was largely attenuated by resveratrol. Immunoprecipitation revealed that SIRT1 physically interacts with FoxO1 in muscle, and this interaction was weakened in 5/6Nx rats. As a consequence, acetylated FoxO1 was increased in muscle of 5/6Nx rats. The application of resveratrol markedly reverses this trend. These data point out that SIRT1 is a key factor for linking renal disease and muscle atrophy. Indeed, both renal dysfunction and muscle atrophy were further aggravated by 5/6Nx in Sirt1+/- mice. Taken together, our data indicate that SIRT1 plays a pivotal role in muscle atrophy in CKD, and FoxO1 might be a substrate of SIRT1 in this process. Furthermore, resveratrol, together with other agonists of SIRT1, may hold great therapeutic potentials for treating CKD and its related muscle atrophy.
Collapse
Affiliation(s)
- Ruiting Wang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Weidong Yuan
- Department of Nephrology, People's Hospital of Haimen District, Nantong, China
| | - Lu Li
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Fei Lu
- Department of Nephrology, People's Hospital of Haimen District, Nantong, China
| | - Lingling Zhang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Haifeng Gong
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xinzhong Huang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
17
|
Yang HW, Oh S, Chung DM, Seo M, Park SJ, Jeon YJ, Byun K, Ryu B. Ishophloroglucin A, Isolated from Ishige okamurae, Alleviates Dexamethasone-Induced Muscle Atrophy through Muscle Protein Metabolism In Vivo. Mar Drugs 2022; 20:280. [PMID: 35621931 PMCID: PMC9147101 DOI: 10.3390/md20050280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
The in vitro capacity of Ishige okamurae extract (IO) to improve impaired muscle function has been previously examined. However, the mechanism underlying IO-mediated muscle protein metabolism and the role of its component, Ishophloroglucin A (IPA), in mice with dexamethasone (Dexa)-induced muscle atrophy remains unknown. In the present study, we evaluated the effect of IO and IPA supplementation on Dexa-induced muscle atrophy by assessing muscle protein metabolism in gastrocnemius and soleus muscles of mice. IO and IPA supplementation improved the Dexa-induced decrease in muscle weight and width, leading to enhanced grip strength. In addition, IO and IPA supplementation regulated impaired protein synthesis (PI3K and Akt) or degradation (muscle-specific ubiquitin ligase muscle RING finger and atrogin-1) by modulating mRNA levels in gastrocnemius and soleus muscles. Additionally, IO and IPA upregulated mRNA levels associated with muscle growth activation (transient receptor potential vanilloid type 4 and adenosine A1 receptor) or inhibition (myostatin and sirtuin 1) in gastrocnemius and soleus muscle tissues of Dexa-induced mice. Collectively, these results suggest that IO and IO-derived IPA can regulate muscle growth through muscle protein metabolism in Dexa-induced muscle atrophy.
Collapse
Affiliation(s)
- Hye-Won Yang
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
| | - Dong-Min Chung
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - Minyoung Seo
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - Shin Jae Park
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Korea
| | - BoMi Ryu
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
| |
Collapse
|
18
|
González-Jamett A, Vásquez W, Cifuentes-Riveros G, Martínez-Pando R, Sáez JC, Cárdenas AM. Oxidative Stress, Inflammation and Connexin Hemichannels in Muscular Dystrophies. Biomedicines 2022; 10:biomedicines10020507. [PMID: 35203715 PMCID: PMC8962419 DOI: 10.3390/biomedicines10020507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 12/16/2022] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of congenital neuromuscular disorders whose clinical signs include myalgia, skeletal muscle weakness, hypotonia, and atrophy that leads to progressive muscle disability and loss of ambulation. MDs can also affect cardiac and respiratory muscles, impairing life-expectancy. MDs in clude Duchenne muscular dystrophy, Emery-Dreifuss muscular dystrophy, facioscapulohumeral muscular dystrophy and limb-girdle muscular dystrophy. These and other MDs are caused by mutations in genes that encode proteins responsible for the structure and function of skeletal muscles, such as components of the dystrophin-glycoprotein-complex that connect the sarcomeric-actin with the extracellular matrix, allowing contractile force transmission and providing stability during muscle contraction. Consequently, in dystrophic conditions in which such proteins are affected, muscle integrity is disrupted, leading to local inflammatory responses, oxidative stress, Ca2+-dyshomeostasis and muscle degeneration. In this scenario, dysregulation of connexin hemichannels seem to be an early disruptor of the homeostasis that further plays a relevant role in these processes. The interaction between all these elements constitutes a positive feedback loop that contributes to the worsening of the diseases. Thus, we discuss here the interplay between inflammation, oxidative stress and connexin hemichannels in the progression of MDs and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
- Correspondence: (A.G.-J.); (A.M.C.)
| | - Walter Vásquez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
| | - Gabriela Cifuentes-Riveros
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
| | - Rafaela Martínez-Pando
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.C.-R.); (R.M.-P.)
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (W.V.); (J.C.S.)
- Correspondence: (A.G.-J.); (A.M.C.)
| |
Collapse
|
19
|
Angelini G, Mura G, Messina G. Therapeutic approaches to preserve the musculature in Duchenne Muscular Dystrophy: The importance of the secondary therapies. Exp Cell Res 2022; 410:112968. [PMID: 34883113 DOI: 10.1016/j.yexcr.2021.112968] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/15/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies (MDs) are heterogeneous diseases, characterized by primary wasting of skeletal muscle, which in severe cases, such as Duchenne Muscular Dystrophy (DMD), leads to wheelchair dependency, respiratory failure, and premature death. Research is ongoing to develop efficacious therapies, particularly for DMD. Most of the efforts, currently focusing on correcting or restoring the primary defect of MDs, are based on gene-addition, exon-skipping, stop codon read-through, and genome-editing. Although promising, most of them revealed several practical limitations. Shared knowledge in the field is that, in order to be really successful, any therapeutic approach has to rely on spared functional muscle tissue, restricting the number of patients eligible for clinical trials to the youngest and less compromised individuals. In line with this, many therapeutic strategies aim to preserve muscle tissue and function. This Review outlines the most interesting and recent studies addressing the secondary outcomes of DMD and how to better deliver the therapeutic agents. In the future, the effective treatment of DMD will likely require combinations of therapies addressing both the primary genetic defect and its consequences.
Collapse
Affiliation(s)
- Giuseppe Angelini
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
20
|
Walayat A, Li Y, Zhang Y, Fu Y, Liu B, Shao XM, Zhang L, Xiao D. Fetal e-cigarette exposure programs a neonatal brain hypoxic-ischemic sensitive phenotype via altering DNA methylation patterns and autophagy signaling pathway. Am J Physiol Regul Integr Comp Physiol 2021; 321:R791-R801. [PMID: 34524928 PMCID: PMC8616627 DOI: 10.1152/ajpregu.00207.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023]
Abstract
Maternal e-cigarette (e-cig) exposure is a pressing perinatal health concern. Emerging evidence reveals its potential adverse impacts on brain development in offspring, yet the underlying mechanisms are poorly understood. The present study tested the hypothesis that fetal e-cig exposure induces an aberrant DNA methylation profile in the developing brain, leading to alteration of autophagic flux signaling and programming of a sensitive phenotype to neonatal hypoxic-ischemic encephalopathy (HIE). Pregnant rats were exposed to chronic intermittent e-cig aerosol. Neonates were examined at the age of 9 days old. Maternal e-cig exposure decreased the body weight and brain weight but enhanced the brain-to-body weight ratio in the neonates. E-cig exposure induced a gender-dependent increase in hypoxic-ischemia-induced brain injury in male neonates associated with enhanced reactive oxygen species (ROS) activity. It differentially altered DNA methyltransferase expression and enhanced both global DNA methylation levels and specific CpG methylation at the autophagy-related gene 5 (ATG5) promoter. In addition, maternal e-cig exposure caused downregulations of ATG5, microtubule-associated protein 1 light chain 3β, and sirtuin 1 expression in neonatal brains. Of importance, knockdown of ATG5 in neonatal pups exaggerated neonatal HIE. In conclusion, the present study reveals that maternal e-cig exposure downregulates autophagy-related gene expression via DNA hypermethylation, leading to programming of a hypoxic-ischemic sensitive phenotype in the neonatal brain.
Collapse
Affiliation(s)
- Andrew Walayat
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Yong Li
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Yanyan Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Yingjie Fu
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Bailin Liu
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xuesi M Shao
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California
| | - Lubo Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Daliao Xiao
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
21
|
Løkken N, Khawajazada T, Storgaard JH, Raaschou-Pedersen D, Christensen ME, Hornsyld TM, Krag T, Ørngreen MC, Vissing J. No effect of resveratrol in patients with mitochondrial myopathy: A cross-over randomized controlled trial. J Inherit Metab Dis 2021; 44:1186-1198. [PMID: 33934389 DOI: 10.1002/jimd.12393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 11/06/2022]
Abstract
Mitochondrial myopathies (MM) are caused by mutations that typically affect genes involved in oxidative phosphorylation. Main symptoms are exercise intolerance and fatigue. Currently, there is no specific treatment for MM. Resveratrol (RSV) is a nutritional supplement that in preclinical studies has been shown to stimulate mitochondrial function. We hypothesized that RSV could improve exercise capacity in patients with MM. The study design was randomized, double-blind, cross-over and placebo-controlled. Eleven patients with genetically verified MM were randomized to receive either 1000 mg/day RSV or placebo (P) for 8 weeks followed by a 4-week washout and then the opposite treatment. Primary outcomes were changes in heart rate (HR) during submaximal cycling exercise and peak oxygen utilization (VO2 max) during maximal exercise. Secondary outcomes included reduction in perceived exertion, changes in lactate concentrations, self-rated function (SF-36) and fatigue scores (FSS), activities of electron transport chain complexes I and IV in mononuclear cells and mitochondrial biomarkers in muscle tissue among others. There were no significant differences in primary and secondary outcomes between treatments. Mean HR changes were -0.3 ± 4.3 (RSV) vs 1.8 ± 5.0 bpm (P), P = .241. Mean VO2 max changes were 0.7 ± 1.4 (RSV) vs -0.2 ± 2.3 mL/min/kg (P), P = .203. The study provides evidence that 1000 mg RSV daily is ineffective in improving exercise capacity in adults with MM. These findings indicate that previous in vitro studies suggesting a therapeutic potential for RSV in MM, do not translate into clinically meaningful effects in vivo.
Collapse
Affiliation(s)
- Nicoline Løkken
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| | - Tahmina Khawajazada
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| | - Jesper Helbo Storgaard
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| | | | - Maja Elling Christensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | | | - Thomas Krag
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| | - Mette C Ørngreen
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Rigshospitalet, University hospital, Copenhagen, Denmark
| |
Collapse
|
22
|
Panza E, Vellecco V, Iannotti FA, Paris D, Manzo OL, Smimmo M, Mitilini N, Boscaino A, de Dominicis G, Bucci M, Di Lorenzo A, Cirino G. Duchenne's muscular dystrophy involves a defective transsulfuration pathway activity. Redox Biol 2021; 45:102040. [PMID: 34174560 PMCID: PMC8246642 DOI: 10.1016/j.redox.2021.102040] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/20/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most frequent X chromosome-linked disease caused by mutations in the gene encoding for dystrophin, leading to progressive and unstoppable degeneration of skeletal muscle tissues. Despite recent advances in the understanding of the molecular processes involved in the pathogenesis of DMD, there is still no cure. In this study, we aim at investigating the potential involvement of the transsulfuration pathway (TSP), and its by-end product namely hydrogen sulfide (H2S), in primary human myoblasts isolated from DMD donors and skeletal muscles of dystrophic (mdx) mice. In myoblasts of DMD donors, we demonstrate that the expression of key genes regulating the H2S production and TSP activity, including cystathionine γ lyase (CSE), cystathionine beta-synthase (CBS), 3 mercaptopyruvate sulfurtransferase (3-MST), cysteine dioxygenase (CDO), cysteine sulfonic acid decarboxylase (CSAD), glutathione synthase (GS) and γ -glutamylcysteine synthetase (γ-GCS) is reduced. Starting from these findings, using Nuclear Magnetic Resonance (NMR) and quantitative Polymerase Chain Reaction (qPCR) we show that the levels of TSP-related metabolites such as methionine, glycine, glutathione, glutamate and taurine, as well as the expression levels of the aforementioned TSP related genes, are significantly reduced in skeletal muscles of mdx mice compared to healthy controls, at both an early (7 weeks) and overt (17 weeks) stage of the disease. Importantly, the treatment with sodium hydrosulfide (NaHS), a commonly used H2S donor, fully recovers the impaired locomotor activity in both 7 and 17 old mdx mice. This is an effect attributable to the reduced expression of pro-inflammatory markers and restoration of autophagy in skeletal muscle tissues. In conclusion, our study uncovers a defective TSP pathway activity in DMD and highlights the role of H2S-donors for novel and safe adjuvant therapy to treat symptoms of DMD.
Collapse
Affiliation(s)
- E Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - V Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - F A Iannotti
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli (NA), Italy
| | - D Paris
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli (NA), Italy
| | - O L Manzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy; Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - M Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - N Mitilini
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - A Boscaino
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - G de Dominicis
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - M Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - A Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - G Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
23
|
Luan P, D'Amico D, Andreux PA, Laurila PP, Wohlwend M, Li H, Imamura de Lima T, Place N, Rinsch C, Zanou N, Auwerx J. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci Transl Med 2021; 13:13/588/eabb0319. [PMID: 33827972 DOI: 10.1126/scitranslmed.abb0319] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/25/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy, and despite advances in genetic and pharmacological disease-modifying treatments, its management remains a major challenge. Mitochondrial dysfunction contributes to DMD, yet the mechanisms by which this occurs remain elusive. Our data in experimental models and patients with DMD show that reduced expression of genes involved in mitochondrial autophagy, or mitophagy, contributes to mitochondrial dysfunction. Mitophagy markers were reduced in skeletal muscle and in muscle stem cells (MuSCs) of a mouse model of DMD. Administration of the mitophagy activator urolithin A (UA) rescued mitophagy in DMD worms and mice and in primary myoblasts from patients with DMD, increased skeletal muscle respiratory capacity, and improved MuSCs' regenerative ability, resulting in the recovery of muscle function and increased survival in DMD mouse models. These data indicate that restoration of mitophagy alleviates symptoms of DMD and suggest that UA may have potential therapeutic applications for muscular dystrophies.
Collapse
Affiliation(s)
- Peiling Luan
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Davide D'Amico
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.,Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pénélope A Andreux
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pirkka-Pekka Laurila
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Hao Li
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
24
|
Nguyen TH, Conotte S, Belayew A, Declèves AE, Legrand A, Tassin A. Hypoxia and Hypoxia-Inducible Factor Signaling in Muscular Dystrophies: Cause and Consequences. Int J Mol Sci 2021; 22:7220. [PMID: 34281273 PMCID: PMC8269128 DOI: 10.3390/ijms22137220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/29/2022] Open
Abstract
Muscular dystrophies (MDs) are a group of inherited degenerative muscle disorders characterized by a progressive skeletal muscle wasting. Respiratory impairments and subsequent hypoxemia are encountered in a significant subgroup of patients in almost all MD forms. In response to hypoxic stress, compensatory mechanisms are activated especially through Hypoxia-Inducible Factor 1 α (HIF-1α). In healthy muscle, hypoxia and HIF-1α activation are known to affect oxidative stress balance and metabolism. Recent evidence has also highlighted HIF-1α as a regulator of myogenesis and satellite cell function. However, the impact of HIF-1α pathway modifications in MDs remains to be investigated. Multifactorial pathological mechanisms could lead to HIF-1α activation in patient skeletal muscles. In addition to the genetic defect per se, respiratory failure or blood vessel alterations could modify hypoxia response pathways. Here, we will discuss the current knowledge about the hypoxia response pathway alterations in MDs and address whether such changes could influence MD pathophysiology.
Collapse
Affiliation(s)
- Thuy-Hang Nguyen
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Stephanie Conotte
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Belayew
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Anne-Emilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium;
| | - Alexandre Legrand
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| |
Collapse
|
25
|
The Interplay of Mitophagy and Inflammation in Duchenne Muscular Dystrophy. Life (Basel) 2021; 11:life11070648. [PMID: 34357020 PMCID: PMC8307817 DOI: 10.3390/life11070648] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a pathogenic disruption of the DYSTROPHIN gene that results in non-functional dystrophin protein. DMD patients experience loss of ambulation, cardiac arrhythmia, metabolic syndrome, and respiratory failure. At the molecular level, the lack of dystrophin in the muscle results in myofiber death, fibrotic infiltration, and mitochondrial dysfunction. There is no cure for DMD, although dystrophin-replacement gene therapies and exon-skipping approaches are being pursued in clinical trials. Mitochondrial dysfunction is one of the first cellular changes seen in DMD myofibers, occurring prior to muscle disease onset and progresses with disease severity. This is seen by reduced mitochondrial function, abnormal mitochondrial morphology and impaired mitophagy (degradation of damaged mitochondria). Dysfunctional mitochondria release high levels of reactive oxygen species (ROS), which can activate pro-inflammatory pathways such as IL-1β and IL-6. Impaired mitophagy in DMD results in increased inflammation and further aggravates disease pathology, evidenced by increased muscle damage and increased fibrosis. This review will focus on the critical interplay between mitophagy and inflammation in Duchenne muscular dystrophy as a pathological mechanism, as well as describe both candidate and established therapeutic targets that regulate these pathways.
Collapse
|
26
|
Qu Z, Zhou S, Li P, Liu C, Yuan B, Zhang S, Liu A. Natural products and skeletal muscle health. J Nutr Biochem 2021; 93:108619. [DOI: 10.1016/j.jnutbio.2021.108619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/26/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022]
|
27
|
Sullivan RT, Lam NT, Haberman M, Beatka MJ, Afzal MZ, Lawlor MW, Strande JL. Cardioprotective effect of nicorandil on isoproterenol induced cardiomyopathy in the Mdx mouse model. BMC Cardiovasc Disord 2021; 21:302. [PMID: 34130633 PMCID: PMC8207777 DOI: 10.1186/s12872-021-02112-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) associated cardiomyopathy is a major cause of morbidity and mortality. In an in vitro DMD cardiomyocyte model, nicorandil reversed stress-induced cell injury through multiple pathways implicated in DMD. We aimed to test the efficacy of nicorandil on the progression of cardiomyopathy in mdx mice following a 10-day treatment protocol. METHODS A subset of mdx mice was subjected to low-dose isoproterenol injections over 5 days to induce a cardiac phenotype and treated with vehicle or nicorandil for 10 days. Baseline and day 10 echocardiograms were obtained to assess cardiac function. At 10 days, cardiac tissue was harvested for further analysis, which included histologic analysis and assessment of oxidative stress. Paired student's t test was used for in group comparison, and ANOVA was used for multiple group comparisons. RESULTS Compared to vehicle treated mice, isoproterenol decreased ejection fraction and fractional shortening on echocardiogram. Nicorandil prevented isoproterenol induced cardiac dysfunction. Isoproterenol increased cardiac fibrosis, which nicorandil prevented. Isoproterenol increased gene expression of NADPH oxidase, which decreased to baseline with nicorandil treatment. Superoxide dismutase 2 protein expression increased in those treated with nicorandil, and xanthine oxidase activity decreased in mice treated with nicorandil during isoproterenol stress compared to all other groups. CONCLUSIONS In conclusion, nicorandil is cardioprotective in mdx mice and warrants continued investigation as a therapy for DMD associated cardiomyopathy.
Collapse
Affiliation(s)
- Rachel T Sullivan
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Ngoc T Lam
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Margaret Haberman
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Margaret J Beatka
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Muhammad Z Afzal
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Michael W Lawlor
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Jennifer L Strande
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| |
Collapse
|
28
|
A Systematic Review on the Role of SIRT1 in Duchenne Muscular Dystrophy. Cells 2021; 10:cells10061380. [PMID: 34205021 PMCID: PMC8229470 DOI: 10.3390/cells10061380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscular disease characterized by progressive muscle degeneration. Life expectancy is between 30 and 50 years, and death is correlated with cardiac or respiratory complications. Currently, there is no cure, so there is a great interest in new pharmacological targets. Sirtuin1 (SIRT1) seems to be a potential target for DMD. In muscle tissue, SIRT1 exerts anti-inflammatory and antioxidant effects. The aim of this study is to summarize all the findings of in vivo and in vitro literature studies about the potential role of SIRT1 in DMD. A systematic literature search was performed according to PRISMA guidelines. Twenty-three articles satisfied the eligibility criteria. It emerged that SIRT1 inhibition led to muscle fragility, while conversely its activation improved muscle function. Additionally, resveratrol, a SIRT1 activator, has brought beneficial effects to the skeletal, cardiac and respiratory muscles by exerting anti-inflammatory activity that leads to reduced myofiber wasting.
Collapse
|
29
|
Bouviere J, Fortunato RS, Dupuy C, Werneck-de-Castro JP, Carvalho DP, Louzada RA. Exercise-Stimulated ROS Sensitive Signaling Pathways in Skeletal Muscle. Antioxidants (Basel) 2021; 10:antiox10040537. [PMID: 33808211 PMCID: PMC8066165 DOI: 10.3390/antiox10040537] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Physical exercise represents a major challenge to whole-body homeostasis, provoking acute and adaptative responses at the cellular and systemic levels. Different sources of reactive oxygen species (ROS) have been described in skeletal muscle (e.g., NADPH oxidases, xanthine oxidase, and mitochondria) and are closely related to the physiological changes induced by physical exercise through the modulation of several signaling pathways. Many signaling pathways that are regulated by exercise-induced ROS generation, such as adenosine monophosphate-activated protein kinase (AMPK), mitogen activated protein kinase (MAPK), nuclear respiratory factor2 (NRF2), and PGC-1α are involved in skeletal muscle responses to physical exercise, such as increased glucose uptake, mitochondriogenesis, and hypertrophy, among others. Most of these adaptations are blunted by antioxidants, revealing the crucial role played by ROS during and after physical exercise. When ROS generation is either insufficient or exacerbated, ROS-mediated signaling is disrupted, as well as physical exercise adaptations. Thus, an understanding the limit between "ROS that can promote beneficial effects" and "ROS that can promote harmful effects" is a challenging question in exercise biology. The identification of new mediators that cause reductive stress and thereby disrupt exercise-stimulated ROS signaling is a trending on this topic and are covered in this current review.
Collapse
Affiliation(s)
- Jessica Bouviere
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Rodrigo S. Fortunato
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Corinne Dupuy
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
| | - Joao Pedro Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Denise P. Carvalho
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Ruy A. Louzada
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Correspondence:
| |
Collapse
|
30
|
The linkage between inflammation and fibrosis in muscular dystrophies: The axis autotaxin-lysophosphatidic acid as a new therapeutic target? J Cell Commun Signal 2021; 15:317-334. [PMID: 33689121 PMCID: PMC8222483 DOI: 10.1007/s12079-021-00610-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MDs) are a diverse group of severe disorders characterized by increased skeletal muscle feebleness. In many cases, respiratory and cardiac muscles are also compromised. Skeletal muscle inflammation and fibrosis are hallmarks of several skeletal muscle diseases, including MDs. Until now, several keys signaling pathways and factors that regulate inflammation and fibrosis have been identified. However, no curative treatments are available. Therefore, it is necessary to find new therapeutic targets to fight these diseases and improve muscle performance. Lysophosphatidic acid (LPA) is an active glycerophospholipid mainly synthesized by the secreted enzyme autotaxin (ATX), which activates six different G protein-coupled receptors named LPA1 to LPA6 (LPARs). In conjunction, they are part of the ATX/LPA/LPARs axis, involved in the inflammatory and fibrotic response in several organs-tissues. This review recapitulates the most relevant aspects of inflammation and fibrosis in MDs. It analyzes experimental evidence of the effects of the ATX/LPA/LPARs axis on inflammatory and fibrotic responses. Finally, we speculate about its potential role as a new therapeutic pharmacological target to treat these diseases.
Collapse
|
31
|
Woodman KG, Coles CA, Lamandé SR, White JD. Resveratrol Promotes Hypertrophy in Wildtype Skeletal Muscle and Reduces Muscle Necrosis and Gene Expression of Inflammatory Markers in Mdx Mice. Molecules 2021; 26:molecules26040853. [PMID: 33561994 PMCID: PMC7915385 DOI: 10.3390/molecules26040853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive fatal neuromuscular disorder with no cure. Therapies to restore dystrophin deficiency have been approved in some jurisdictions but long-term effectiveness is yet to be established. There is a need to develop alternative strategies to treat DMD. Resveratrol is a nutraceutical with anti-inflammatory properties. Previous studies have shown high doses (100–400 mg/kg bodyweight/day) benefit mdx mice. We treated 4-week-old mdx and wildtype mice with a lower dose of resveratrol (5 mg/kg bodyweight/day) for 15 weeks. Voluntary exercise was used to test if a lower dosage than previously tested could reduce exercise-induced damage where a greater inflammatory infiltrate is present. We found resveratrol promoted skeletal muscle hypertrophy in wildtype mice. In dystrophic muscle, resveratrol reduced exercise-induced muscle necrosis. Gene expression of immune cell markers, CD86 and CD163 were reduced; however, signalling targets associated with resveratrol’s mechanism of action including Sirt1 and NF-κB were unchanged. In conclusion, a lower dose of resveratrol compared to the dosage used by other studies reduced necrosis and gene expression of inflammatory cell markers in dystrophic muscle suggesting it as a therapeutic candidate for treating DMD.
Collapse
Affiliation(s)
- Keryn G. Woodman
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia;
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, VIC 3010, Australia;
- Department of Genetics, Yale University, New Haven, CT 06510, USA
- Correspondence: (K.G.W.); (C.A.C.); Tel.: +1-203-737-1091 (K.G.W.); +61-3-9936-6021(C.A.C.)
| | - Chantal A. Coles
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia;
- Correspondence: (K.G.W.); (C.A.C.); Tel.: +1-203-737-1091 (K.G.W.); +61-3-9936-6021(C.A.C.)
| | - Shireen R. Lamandé
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia;
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jason D. White
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, VIC 3010, Australia;
- Office of the Pro Vice Chancellor Research and Innovation, Charles Sturt University, Wagga, NSW 2678, Australia
| |
Collapse
|
32
|
Singh K, Randhwa G, Salloum FN, Grider JR, Murthy KS. Decreased smooth muscle function, peristaltic activity, and gastrointestinal transit in dystrophic (mdx) mice. Neurogastroenterol Motil 2021; 33:e13968. [PMID: 32789934 DOI: 10.1111/nmo.13968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is characterized by the lack of dystrophin in skeletal, cardiac, and smooth muscle. Slow colonic transit and constipation are common in DMD patients and animal models of DMD. However, the cause of this hypocontractility and the expression of contractile proteins in smooth muscle are unknown. The aim of the study was to investigate the expression of contractile proteins in the colonic smooth muscle and the function of the colon in control and mdx mice. METHODS Muscle contraction was measured in muscle strips and isolated muscle cells. Peristaltic activity was measured in ex vivo preparations by spatiotemporal mapping, and gastrointestinal (GI) transit in vivo was measured by the distribution of fluorescent marker along the intestine and colon. mRNA expression of contractile proteins smoothelin, caldesmon, calponin, and tropomyosin was measured by qRT-PCR. RESULTS Expression of mRNA for contractile proteins was decreased in colonic smooth muscle of mdx mice compared with control. Contraction in response to acetylcholine and KCl was decreased in colonic muscle strips and in isolated muscle cells of mdx mice. Distension of ex vivo colons with Krebs buffer induced peristalsis in both control and mdx mice; however, significantly fewer full peristaltic waves were recorded in the colons of mdx mice. GI transit was also inhibited in mdx mice. CONCLUSION AND INFERENCES The data indicate that the lack of dystrophin causes decrease in colonic smooth muscle contractility, peristalsis, and GI transit and provides the basis for analysis of mechanisms involved in smooth muscle dysfunction in DMD.
Collapse
Affiliation(s)
- Kulpreet Singh
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Gurpreet Randhwa
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Fadi N Salloum
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - John R Grider
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Karnam S Murthy
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
33
|
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: 21] [Impact Index Per Article: 7.0] [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
|
34
|
Jia Z, Yang X, Liu K. Treatment of cattle oocytes with C-type natriuretic peptide before in vitro maturation enhances oocyte mitochondrial function. Anim Reprod Sci 2020; 225:106685. [PMID: 33388612 DOI: 10.1016/j.anireprosci.2020.106685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 11/30/2022]
Abstract
The present study was conducted to evaluate the effects in vitro on oocyte mitochondrial function of C-type natriuretic peptide (CNP) when treatments were imposed before in vitro maturation (IVM). Immature oocytes were either directly matured in vitro for 24 h (Control, no pre-IVM), or cultured in basic medium not supplemented or supplemented with CNP (100 nM) (Control pre-IVM and CNP pre-IVM, respectively) for 6 h, followed by IVM for 24 h. The results indicated treatment with CNP before IVM affected patterns of distribution of mitochondria, increased the mitochondrial content, membrane potential, and decreased the ROS content in cattle oocytes before and after IVM. Furthermore, treatment of immature cattle oocytes with CNP before IVM induced marked increases in the relative abundance of mRNA transcripts and proteins related to mitochondria development and antioxidative defense mechanisms. Treatment with CNP before oocyte IVM also resulted in an enhanced relative abundance of sirtuin-1 (SIRT1) mRNA transcript in cattle oocytes. Taken together, these results provide evidence that treatment of cattle oocytes with CNP before IVM improved mitochondrial function and antioxidant defense mechanisms in cattle oocytes. Findings in the present study provide insights into the potential mechanisms by which CNP has positive effects on oocyte cytoplasmic organelles, specifically mitochondria.
Collapse
Affiliation(s)
- Zhenwei Jia
- College of Animal Science and Technology, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, 028000, PR China.
| | - Xinyu Yang
- College of Animal Science and Technology, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, 028000, PR China
| | - Kai Liu
- College of Animal Science and Technology, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, 028000, PR China; Inner Mongolia Engineering Technology Research Center for the Beef Cattle Disease Prevention and Control, Tongliao, Inner Mongolia, 028000, PR China
| |
Collapse
|
35
|
Rybalka E, Timpani CA, Debruin DA, Bagaric RM, Campelj DG, Hayes A. The Failed Clinical Story of Myostatin Inhibitors against Duchenne Muscular Dystrophy: Exploring the Biology behind the Battle. Cells 2020; 9:E2657. [PMID: 33322031 PMCID: PMC7764137 DOI: 10.3390/cells9122657] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
Myostatin inhibition therapy has held much promise for the treatment of muscle wasting disorders. This is particularly true for the fatal myopathy, Duchenne Muscular Dystrophy (DMD). Following on from promising pre-clinical data in dystrophin-deficient mice and dogs, several clinical trials were initiated in DMD patients using different modality myostatin inhibition therapies. All failed to show modification of disease course as dictated by the primary and secondary outcome measures selected: the myostatin inhibition story, thus far, is a failed clinical story. These trials have recently been extensively reviewed and reasons why pre-clinical data collected in animal models have failed to translate into clinical benefit to patients have been purported. However, the biological mechanisms underlying translational failure need to be examined to ensure future myostatin inhibitor development endeavors do not meet with the same fate. Here, we explore the biology which could explain the failed translation of myostatin inhibitors in the treatment of DMD.
Collapse
Affiliation(s)
- Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Cara A. Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Danielle A. Debruin
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Ryan M. Bagaric
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Dean G. Campelj
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Alan Hayes
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
- Department of Medicine—Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, 3021 Victoria, Australia
| |
Collapse
|
36
|
Bronisz-Budzyńska I, Kozakowska M, Podkalicka P, Kachamakova-Trojanowska N, Łoboda A, Dulak J. The role of Nrf2 in acute and chronic muscle injury. Skelet Muscle 2020; 10:35. [PMID: 33287890 PMCID: PMC7722332 DOI: 10.1186/s13395-020-00255-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) is considered as a master cytoprotective factor regulating the expression of genes encoding anti-oxidant, anti-inflammatory, and detoxifying proteins. The role of Nrf2 in the pathophysiology of skeletal muscles has been evaluated in different experimental models, however, due to inconsistent data, we aimed to investigate how Nrf2 transcriptional deficiency (Nrf2tKO) affects muscle functions both in an acute and chronic injury. The acute muscle damage was induced in mice of two genotypes-WT and Nrf2tKO mice by cardiotoxin (CTX) injection. To investigate the role of Nrf2 in chronic muscle pathology, mdx mice that share genetic, biochemical, and histopathological features with Duchenne muscular dystrophy (DMD) were crossed with mice lacking transcriptionally active Nrf2 and double knockouts (mdx/Nrf2tKO) were generated. To worsen the dystrophic phenotype, the analysis of disease pathology was also performed in aggravated conditions, by applying a long-term treadmill test. We have observed slightly increased muscle damage in Nrf2tKO mice after CTX injection. Nevertheless, transcriptional ablation of Nrf2 in mdx mice did not significantly aggravate the most deleterious, pathological hallmarks of DMD related to degeneration, inflammation, fibrotic scar formation, angiogenesis, and the number and proliferation of satellite cells in non-exercised conditions. On the other hand, upon chronic exercises, the degeneration and inflammatory infiltration of the gastrocnemius muscle, but not the diaphragm, turned to be increased in Nrf2tKOmdx in comparison to mdx mice. In conclusion, the lack of transcriptionally active Nrf2 influences moderately muscle pathology in acute CTX-induced muscle injury and chronic DMD mouse model, without affecting muscle functionality. Hence, in general, we demonstrated that the deficiency of Nrf2 transcriptional activity has no profound impact on muscle pathology in various models of muscle injury.
Collapse
Affiliation(s)
- Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | | | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
37
|
van Westering TLE, Johansson HJ, Hanson B, Coenen-Stass AML, Lomonosova Y, Tanihata J, Motohashi N, Yokota T, Takeda S, Lehtiö J, Wood MJA, El Andaloussi S, Aoki Y, Roberts TC. Mutation-independent Proteomic Signatures of Pathological Progression in Murine Models of Duchenne Muscular Dystrophy. Mol Cell Proteomics 2020; 19:2047-2068. [PMID: 32994316 PMCID: PMC7710136 DOI: 10.1074/mcp.ra120.002345] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
The absence of the dystrophin protein in Duchenne muscular dystrophy (DMD) results in myofiber fragility and a plethora of downstream secondary pathologies. Although a variety of experimental therapies are in development, achieving effective treatments for DMD remains exceptionally challenging, not least because the pathological consequences of dystrophin loss are incompletely understood. Here we have performed proteome profiling in tibialis anterior muscles from two murine DMD models (mdx and mdx52) at three ages (8, 16, and 80 weeks of age), all n = 3. High-resolution isoelectric focusing liquid chromatography-tandem MS (HiRIEF-LC-MS/MS) was used to quantify the expression of 4974 proteins across all 27 samples. The two dystrophic models were found to be highly similar, whereas multiple proteins were differentially expressed relative to WT (C57BL/6) controls at each age. Furthermore, 1795 proteins were differentially expressed when samples were pooled across ages and dystrophic strains. These included numerous proteins associated with the extracellular matrix and muscle function that have not been reported previously. Pathway analysis revealed multiple perturbed pathways and predicted upstream regulators, which together are indicative of cross-talk between inflammatory, metabolic, and muscle growth pathways (e.g. TNF, INFγ, NF-κB, SIRT1, AMPK, PGC-1α, PPARs, ILK, and AKT/PI3K). Upregulation of CAV3, MVP and PAK1 protein expression was validated in dystrophic muscle by Western blot. Furthermore, MVP was upregulated during, but not required for, the differentiation of C2C12 myoblasts suggesting that this protein may affect muscle regeneration. This study provides novel insights into mutation-independent proteomic signatures characteristic of the dystrophic phenotype and its progression with aging.
Collapse
Affiliation(s)
| | - Henrik J Johansson
- Department of Oncology/Pathology, Cancer Proteomics Mass Spectrometry, SciLifeLab Stockholm, Karolinska Institutet, Stockholm, Sweden
| | - Britt Hanson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Yulia Lomonosova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Toshifumi Yokota
- Department of Medical, Genetics, School of Human Development Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Janne Lehtiö
- Department of Oncology/Pathology, Cancer Proteomics Mass Spectrometry, SciLifeLab Stockholm, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; MDUK Oxford Neuromuscular Centre, Oxford, UK
| | | | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan.
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
38
|
Segatto M, Szokoll R, Fittipaldi R, Bottino C, Nevi L, Mamchaoui K, Filippakopoulos P, Caretti G. BETs inhibition attenuates oxidative stress and preserves muscle integrity in Duchenne muscular dystrophy. Nat Commun 2020; 11:6108. [PMID: 33257646 PMCID: PMC7705749 DOI: 10.1038/s41467-020-19839-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/31/2020] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects 1 in 3500 live male births. To date, there is no effective cure for DMD, and the identification of novel molecular targets involved in disease progression is important to design more effective treatments and therapies to alleviate DMD symptoms. Here, we show that protein levels of the Bromodomain and extra-terminal domain (BET) protein BRD4 are significantly increased in the muscle of the mouse model of DMD, the mdx mouse, and that pharmacological inhibition of the BET proteins has a beneficial outcome, tempering oxidative stress and muscle damage. Alterations in reactive oxygen species (ROS) metabolism are an early event in DMD onset and they are tightly linked to inflammation, fibrosis, and necrosis in skeletal muscle. By restoring ROS metabolism, BET inhibition ameliorates these hallmarks of the dystrophic muscle, translating to a beneficial effect on muscle function. BRD4 direct association to chromatin regulatory regions of the NADPH oxidase subunits increases in the mdx muscle and JQ1 administration reduces BRD4 and BRD2 recruitment at these regions. JQ1 treatment reduces NADPH subunit transcript levels in mdx muscles, isolated myofibers and DMD immortalized myoblasts. Our data highlight novel functions of the BET proteins in dystrophic skeletal muscle and suggest that BET inhibitors may ameliorate the pathophysiology of DMD.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy.,Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche (Is), Italy
| | - Roberta Szokoll
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Raffaella Fittipaldi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Cinzia Bottino
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Lorenzo Nevi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, U974, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Panagis Filippakopoulos
- Structural Genomics Consortium, Old Road Campus Research Building, Nuffield Department of Medicine, Oxford, OX3 7DQ, UK
| | - Giuseppina Caretti
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
39
|
Kawamura K, Fukumura S, Nikaido K, Tachi N, Kozuka N, Seino T, Hatakeyama K, Mori M, Ito YM, Takami A, Hinotsu S, Kuno A, Kawasaki Y, Horio Y, Tsutsumi H. Resveratrol improves motor function in patients with muscular dystrophies: an open-label, single-arm, phase IIa study. Sci Rep 2020; 10:20585. [PMID: 33239684 PMCID: PMC7688653 DOI: 10.1038/s41598-020-77197-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Muscular dystrophies (MDs) are inherited disorders characterized by progressive muscle weakness. Previously, we have shown that resveratrol (3,5,4′-trihydroxy-trans-stilbene), an antioxidant and an activator of the protein deacetylase SIRT1, decreases muscular and cardiac oxidative damage and improves pathophysiological conditions in animal MD models. To determine whether resveratrol provides therapeutic benefits to patients with MDs, an open-label, single-arm, phase IIa trial of resveratrol was conducted in 11 patients with Duchenne, Becker or Fukuyama MD. The daily dose of resveratrol was 500 mg/day, which was increased every 8 weeks to 1000 and then 1500 mg/day. Primary outcomes were motor function, evaluated by a motor function measure (MFM) scale, muscular strength, monitored with quantitative muscle testing (QMT), and serum creatine kinase (CK) levels. Adverse effects and tolerability were evaluated as secondary outcomes. Despite the advanced medical conditions of the patients, the mean MFM scores increased significantly from 34.6 to 38.4 after 24 weeks of medication. A twofold increase was found in the mean QMT scores of scapula elevation and shoulder abduction. Mean CK levels decreased considerably by 34%. Diarrhoea and abdominal pain was noted in six and three patients, respectively. Resveratrol may provide some benefit to MD patients.
Collapse
Affiliation(s)
- Kentaro Kawamura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Koki Nikaido
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Nobutada Tachi
- Faculty of Health Science, Hokkaido Chitose College of Rehabilitation, Chitose, 066-0055, Japan
| | - Naoki Kozuka
- Department of Physical Therapy, Sapporo Medical University School of Health Sciences, Sapporo, 060-8556, Japan
| | - Tsugumi Seino
- Department of Physical Therapy, Sapporo Medical University School of Health Sciences, Sapporo, 060-8556, Japan
| | - Kingya Hatakeyama
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Mitsuru Mori
- Faculty of Health Science, Hokkaido Chitose College of Rehabilitation, Chitose, 066-0055, Japan
| | - Yoichi M Ito
- Biostatistics Division, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Akiyoshi Takami
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, 480-1195, Japan
| | - Shiro Hinotsu
- Department of Biostatistics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Yukihiko Kawasaki
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan.
| | - Hiroyuki Tsutsumi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan.
| |
Collapse
|
40
|
Ashrafizadeh M, Najafi M, Orouei S, Zabolian A, Saleki H, Azami N, Sharifi N, Hushmandi K, Zarrabi A, Ahn KS. Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities. Biomedicines 2020; 8:E261. [PMID: 32752069 PMCID: PMC7460084 DOI: 10.3390/biomedicines8080261] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Resveratrol (Res) is a well-known natural product that can exhibit important pharmacological activities such as antioxidant, anti-diabetes, anti-tumor, and anti-inflammatory. An evaluation of its therapeutic effects demonstrates that this naturally occurring bioactive compound can target different molecular pathways to exert its pharmacological actions. Transforming growth factor-beta (TGF-β) is an important molecular pathway that is capable of regulating different cellular mechanisms such as proliferation, migration, and angiogenesis. TGF-β has been reported to be involved in the development of disorders such as diabetes, cancer, inflammatory disorders, fibrosis, cardiovascular disorders, etc. In the present review, the relationship between Res and TGF-β has been investigated. It was noticed that Res can inhibit TGF-β to suppress the proliferation and migration of cancer cells. In addition, Res can improve fibrosis by reducing inflammation via promoting TGF-β down-regulation. Res has been reported to be also beneficial in the amelioration of diabetic complications via targeting the TGF-β signaling pathway. These topics are discussed in detail in this review to shed light on the protective effects of Res mediated via the modulation of TGF-β signaling.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Sima Orouei
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Negar Azami
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Negin Sharifi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956 Istanbul, Turkey
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
41
|
El kiki SM, Omran MM, Mansour HH, Hasan HF. Metformin and/or low dose radiation reduces cardiotoxicity and apoptosis induced by cyclophosphamide through SIRT-1/SOD and BAX/Bcl-2 pathways in rats. Mol Biol Rep 2020; 47:5115-5126. [DOI: 10.1007/s11033-020-05582-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/10/2020] [Indexed: 12/27/2022]
|
42
|
Liu CW, Huang CC, Hsu CF, Li TH, Tsai YL, Lin MW, Tsai HC, Huang SF, Yang YY, Hsieh YC, Lee TY, Tsai CY, Huang YH, Hou MC, Lin HC. SIRT1-dependent mechanisms and effects of resveratrol for amelioration of muscle wasting in NASH mice. BMJ Open Gastroenterol 2020; 7:bmjgast-2020-000381. [PMID: 32371503 PMCID: PMC7228468 DOI: 10.1136/bmjgast-2020-000381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/21/2022] Open
Abstract
Background In non-alcoholic steatohepatitis (NASH), muscle wasting was an aggravating factor for the progression of hepatic steatosis. This study explores the potential benefits of chronic treatment with resveratrol, a strong activator of SIRT1 on the muscle wasting of NASH mice. Methods In vivo and in vitro study, we evaluate the SIRT1-dependent mechanisms and effects of resveratrol administration for 6 weeks with high-fat-methionine and choline deficient diet-induced NASH mice and palmitate-pretreated C2C12 myoblast cells. Results Resveratrol treatment improved grip strength and muscle mass of limbs, increased running distance and time on exercise wheels in NASH mice. There is a negative correlation between muscular SIRT1 activity and 3-nitrotyrosine levels of NASH and NASH-resv mice. The SIRT1-dependent effect of muscle wasting was associated with the suppression of oxidative stress, upregulation of antioxidants, inhibition of protein degradation, activation of autophagy, suppression of apoptotic activity, upregulation of lipolytic genes and the reduction of fatty infiltration in limb muscles of NASH mice. In vitro, resveratrol alleviated palmitate acid-induced oxidative stress, lipid deposition, autophagy dysfunction, apoptotic signals, and subsequently reduced fusion index and myotube formation of C2C12 cells. The beneficial effects of resveratrol were abolished by EX527. Conclusions Our study suggests that chronic resveratrol treatment is a potential strategy for amelioration of hepatic steatosis and muscle wasting in NASH mouse model.
Collapse
Affiliation(s)
- Chih-Wei Liu
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, Taipei, Taiwan
| | - Chia-Chang Huang
- Institute of Clinical Medicine, Taipei, Taiwan.,Division of Clinical Skills Center, Department of Medical Education, Taipei Veterans General Hospital, Taoyuan, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chien-Fu Hsu
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Hao Li
- Institute of Clinical Medicine, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Lien Tsai
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Wei Lin
- Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Institute of Public Health, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hung-Cheng Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shiang-Fen Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Infection Disease, Taipei, Taiwan
| | - Ying-Ying Yang
- Institute of Clinical Medicine, Taipei, Taiwan .,Division of Clinical Skills Center, Department of Medical Education, Taipei Veterans General Hospital, Taoyuan, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Yun-Cheng Hsieh
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Institute of Clinical Medicine, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Ming-Chih Hou
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Han-Chieh Lin
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| |
Collapse
|
43
|
Suntar I, Sureda A, Belwal T, Sanches Silva A, Vacca RA, Tewari D, Sobarzo-Sánchez E, Nabavi SF, Shirooie S, Dehpour AR, Xu S, Yousefi B, Majidinia M, Daglia M, D'Antona G, Nabavi SM. Natural products, PGC-1 α , and Duchenne muscular dystrophy. Acta Pharm Sin B 2020; 10:734-745. [PMID: 32528825 PMCID: PMC7276681 DOI: 10.1016/j.apsb.2020.01.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/14/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a transcriptional coactivator that binds to a diverse range of transcription factors. PPARγ coactivator 1 (PGC-1) coactivators possess an extensive range of biological effects in different tissues, and play a key part in the regulation of the oxidative metabolism, consequently modulating the production of reactive oxygen species, autophagy, and mitochondrial biogenesis. Owing to these findings, a large body of studies, aiming to establish the role of PGC-1 in the neuromuscular system, has shown that PGC-1 could be a promising target for therapies targeting neuromuscular diseases. Among these, some evidence has shown that various signaling pathways linked to PGC-1α are deregulated in muscular dystrophy, leading to a reduced capacity for mitochondrial oxidative phosphorylation and increased reactive oxygen species (ROS) production. In the light of these results, any intervention aimed at activating PGC-1 could contribute towards ameliorating the progression of muscular dystrophies. PGC-1α is influenced by different patho-physiological/pharmacological stimuli. Natural products have been reported to display modulatory effects on PPARγ activation with fewer side effects in comparison to synthetic drugs. Taken together, this review summarizes the current knowledge on Duchenne muscular dystrophy, focusing on the potential effects of natural compounds, acting as regulators of PGC-1α.
Collapse
Key Words
- AAV, adeno-associated virus
- AMP, adenosine monophosphate
- AMPK, 5′ adenosine monophosphate-activated protein kinase
- ASO, antisense oligonucleotides
- ATF2, activating transcription factor 2
- ATP, adenosine triphosphate
- BMD, Becker muscular dystrophy
- COPD, chronic obstructive pulmonary disease
- CREB, cyclic AMP response element-binding protein
- CnA, calcineurin a
- DAGC, dystrophin-associated glycoprotein complex
- DGC, dystrophin–glycoprotein complex
- DMD, Duchenne muscular dystrophy
- DRP1, dynamin-related protein 1
- DS, Down syndrome
- ECM, extracellular matrix
- EGCG, epigallocatechin-3-gallate
- ERRα, estrogen-related receptor alpha
- FDA, U. S. Food and Drug Administration
- FGF, fibroblast growth factor
- FOXO1, forkhead box class-O1
- GABP, GA-binding protein
- GPX, glutathione peroxidase
- GSK3b, glycogen synthase kinase 3b
- HCT, hydrochlorothiazide
- HDAC, histone deacetylase
- HIF-1α, hypoxia-inducible factors
- IL, interleukin
- LDH, lactate dehydrogenase
- MCP-1, monocyte chemoattractant protein-1
- MD, muscular dystrophy
- MEF2, myocyte enhancer factor 2
- MSCs, mesenchymal stem cells
- Mitochondrial oxidative phosphorylation
- Muscular dystrophy
- MyoD, myogenic differentiation
- NADPH, nicotinamide adenine dinucleotide phosphate
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NMJ, neuromuscular junctions
- NO, nitric oxide
- NOS, NO synthase
- Natural product
- PDGF, platelet derived growth factor
- PGC-1, peroxisome proliferator-activated receptor γ coactivator 1
- PPARγ activation
- PPARγ, peroxisome proliferator-activated receptor γ
- Peroxisome proliferator-activated receptor γ coactivator 1α
- ROS, reactive oxygen species
- Reactive oxygen species
- SIRT1, silent mating type information regulation 2 homolog 1
- SOD, superoxide dismutase
- SPP1, secreted phosphoprotein 1
- TNF-α, tumor necrosis factor-α
- UCP, uncoupling protein
- VEGF, vascular endothelial growth factor
- cGMP, cyclic guanosine monophosphate
- iPSCs, induced pluripotent stem cells
- p38 MAPK, p38 mitogen-activated protein kinase
Collapse
|
44
|
Resveratrol reduces oxidative damage and inflammation in mice infected with Trichinella spiralis. J Helminthol 2020; 94:e140. [PMID: 32238206 DOI: 10.1017/s0022149x20000206] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Trichinellosis is a serious food-borne zoonotic infection of cosmopolitan distribution. Currently, treatment for trichinellosis is far from ideal. Given the important role of oxidative stress and immune-mediated inflammation in the pathogenesis of trichinellosis, this study was designed to evaluate the possible protective effects of resveratrol (RSV) during the intestinal and muscular phases of Trichinella spiralis infection in mice. The oral administration of RSV at a dose of 20 mg/kg once daily for two weeks resulted in significant reductions in both adult and larval counts; significant improvements in the redox status of the small intestine and muscles; a significant reduction in interleukin 4, pentraxin 3 and vascular endothelial growth factor expression; and the mitigation of intestinal and muscular inflammation. In conclusion, this study identifies RSV as a promising agent for the treatment of experimental trichinellosis, and more studies in experimental animals and humans are worth consideration.
Collapse
|
45
|
Razzoli M, Lindsay A, Law ML, Chamberlain CM, Southern WM, Berg M, Osborn J, Engeland WC, Metzger JM, Ervasti JM, Bartolomucci A. Social stress is lethal in the mdx model of Duchenne muscular dystrophy. EBioMedicine 2020; 55:102700. [PMID: 32192914 PMCID: PMC7251247 DOI: 10.1016/j.ebiom.2020.102700] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by the loss of dystrophin. Severe and ultimately lethal, DMD progresses relatively slowly in that patients become wheelchair bound only around age twelve with a survival expectancy reaching the third decade of life. Methods The mildly-affected mdx mouse model of DMD, and transgenic DysΔMTB-mdx and Fiona-mdx mice expressing dystrophin or utrophin, respectively, were exposed to either mild (scruffing) or severe (subordination stress) stress paradigms and profiled for their behavioral and physiological responses. A subgroup of mdx mice exposed to subordination stress were pretreated with the beta-blocker metoprolol. Findings Subordination stress caused lethality in ∼30% of mdx mice within 24 h and ∼70% lethality within 48 h, which was not rescued by metoprolol. Lethality was associated with heart damage, waddling gait and hypo-locomotion, as well as marked up-regulation of the hypothalamus-pituitary-adrenocortical axis. A novel cardiovascular phenotype emerged in mdx mice, in that scruffing caused a transient drop in arterial pressure, while subordination stress caused severe and sustained hypotension with concurrent tachycardia. Transgenic expression of dystrophin or utrophin in skeletal muscle protected mdx mice from scruffing and social stress-induced responses including mortality. Interpretation We have identified a robust new stress phenotype in the otherwise mildly affected mdx mouse that suggests relatively benign handling may impact the outcome of behavioural experiments, but which should also expedite the knowledge-based therapy development for DMD. Funding Greg Marzolf Jr. Foundation, Summer's Wish Fund, NIAMS, Muscular Dystrophy Association, University of Minnesota and John and Cheri Gunvalson Trust.
Collapse
Affiliation(s)
- Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Angus Lindsay
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle L Law
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William M Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Madeleine Berg
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - John Osborn
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William C Engeland
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States.
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States.
| |
Collapse
|
46
|
Saclier M, Bonfanti C, Antonini S, Angelini G, Mura G, Zanaglio F, Taglietti V, Romanello V, Sandri M, Tonelli C, Petroni K, Cassano M, Messina G. Nutritional intervention with cyanidin hinders the progression of muscular dystrophy. Cell Death Dis 2020; 11:127. [PMID: 32071288 PMCID: PMC7028923 DOI: 10.1038/s41419-020-2332-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/25/2022]
Abstract
Muscular Dystrophies are severe genetic diseases due to mutations in structural genes, characterized by progressive muscle wasting that compromises patients' mobility and respiratory functions. Literature underlined oxidative stress and inflammation as key drivers of these pathologies. Interestingly among different myofiber classes, type I fibers display a milder dystrophic phenotype showing increased oxidative metabolism. This work shows the benefits of a cyanidin-enriched diet, that promotes muscle fiber-type switch and reduced inflammation in dystrophic alpha-sarcoglyan (Sgca) null mice having, as a net outcome, morphological and functional rescue. Notably, this benefit is achieved also when the diet is administered in dystrophic animals when the signs of the disease are seriously evident. Our work provides compelling evidence that a cyanidin-rich diet strongly delays the progression of muscular dystrophies, paving the way for a combinatorial approach where nutritional-based reduction of muscle inflammation and oxidative stress facilitate the successful perspectives of definitive treatments.
Collapse
Affiliation(s)
- Marielle Saclier
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Chiara Bonfanti
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Stefania Antonini
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Giuseppe Angelini
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Federica Zanaglio
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Valentina Taglietti
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Vanina Romanello
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Chiara Tonelli
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Katia Petroni
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Marco Cassano
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
47
|
Polyphenols and their potential role in preventing skeletal muscle atrophy. Nutr Res 2020; 74:10-22. [DOI: 10.1016/j.nutres.2019.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/18/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022]
|
48
|
Patel A, Vendrell-Gonzalez S, Haas G, Marcinczyk M, Ziemkiewicz N, Talovic M, Fisher JS, Garg K. Regulation of Myogenic Activity by Substrate and Electrical Stimulation In Vitro. Biores Open Access 2019; 8:129-138. [PMID: 31367477 PMCID: PMC6664826 DOI: 10.1089/biores.2019.0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle has a remarkable regenerative capacity in response to mild injury. However, when muscle is severely injured, muscle regeneration is impaired due to the loss of muscle-resident stem cells, known as satellite cells. Fibrotic tissue, primarily comprising collagen I (COL), is deposited with this critical loss of muscle. In recent studies, supplementation of laminin (LM)-111 has been shown to improve skeletal muscle regeneration in several models of disease and injury. Additionally, electrical stimulation (E-stim) has been investigated as a possible rehabilitation therapy to improve muscle's functional recovery. This study investigated the role of E-stim and substrate in regulating myogenic response. C2C12 myoblasts were allowed to differentiate into myotubes on COL- and LM-coated polydimethylsiloxane molds. The myotubes were subjected to E-stim and compared with nonstimulated controls. While E-stim resulted in increased myogenic activity, irrespective of substrate, LM supported increased proliferation and uniform distribution of C2C12 myoblasts. In addition, C2C12 myoblasts cultured on LM showed higher Sirtuin 1, mammalian target of rapamycin, desmin, nitric oxide, and vascular endothelial growth factor expression. Taken together, these results suggest that an LM substrate is more conducive to myoblast growth and differentiation in response to E-stim in vitro.
Collapse
Affiliation(s)
- Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Sara Vendrell-Gonzalez
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Jonathan S Fisher
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
49
|
Fujiwara D, Iwahara N, Sebori R, Hosoda R, Shimohama S, Kuno A, Horio Y. SIRT1 deficiency interferes with membrane resealing after cell membrane injury. PLoS One 2019; 14:e0218329. [PMID: 31242212 PMCID: PMC6594621 DOI: 10.1371/journal.pone.0218329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/30/2019] [Indexed: 01/07/2023] Open
Abstract
Activation of SIRT1, an NAD+-dependent protein deacetylase, ameliorates muscular pathophysiology of δ-sarcoglycan-deficient TO-2 hamsters and dystrophin-deficient mdx mice. We found that SIRT1 was highly expressed beneath the cellular membranes of muscle cells. To elucidate functional roles of SIRT1 on muscles, skeletal muscle-specific SIRT1 knockout mice (SIRT1-MKO) were generated. SIRT1-MKO mice showed muscular pathology similar to mild muscular dystrophies with increased numbers of centrally nucleated small myofibers and decreased numbers of middle-sized (2000–3001 μm2) myofibers compared to those of wild-type (WT) mice. Accordingly, SIRT1-MKO mice showed significantly decreased exercise capacity in treadmill and inverted hanging tests with higher levels of serum creatine kinase activities compared with those in WT mice. Evans blue dye uptake after exercise was greater in the muscles of SIRT1-MKO than those of WT mice, suggesting membrane fragility in SIRT1-MKO mice. Because SIRT1 was dominantly localized beneath the membranes of muscular cells, SIRT1 may have a new role in the membranes. We found that levels of fluorescent FM1-43 dye intake after laser-induced membrane disruption in C2C12 cells were significantly increased by SIRT1 inhibitors or Sirt1-siRNA compared with those of control cells. Inhibition of SIRT1 or SIRT1-knockdown severely disturbed the dynamic aggregation of membrane vesicles under the injured site but did not affect expression levels of membrane repair proteins. These data suggested that SIRT1 had a critical role in the resealing of membrane-ruptured muscle cells, which could affect phenotypes of SIRT1-MKO mice. To our knowledge, this report is the first to demonstrate that SIRT1 affected plasma-membrane repair mechanisms.
Collapse
Affiliation(s)
- Daisuke Fujiwara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Neurology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Rio Sebori
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shun Shimohama
- Department of Neurology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
- * E-mail:
| |
Collapse
|
50
|
Vignier N, Chatzifrangkeskou M, Morales Rodriguez B, Mericskay M, Mougenot N, Wahbi K, Bonne G, Muchir A. Rescue of biosynthesis of nicotinamide adenine dinucleotide protects the heart in cardiomyopathy caused by lamin A/C gene mutation. Hum Mol Genet 2019; 27:3870-3880. [PMID: 30053027 DOI: 10.1093/hmg/ddy278] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 01/07/2023] Open
Abstract
Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is an anatomic and pathologic condition associated with muscle and electrical dysfunction of the heart, often leading to heart failure-related disability. There is currently no specific therapy available for patients that target the molecular pathophysiology of LMNA cardiomyopathy. Recent studies suggested that nicotinamide adenine dinucleotide (NAD+) cellular content could be a critical determinant for heart function. Biosynthesis of NAD+ from vitamin B3 (known as salvage pathways) is the primary source of NAD+. We showed here that NAD+ salvage pathway was altered in the heart of mouse and human carrying LMNA mutation, leading to an alteration of one of NAD+ co-substrate enzymes, PARP-1. Oral administration of nicotinamide riboside, a natural NAD+ precursor and a pyridine-nucleoside form of vitamin B3, leads to a marked improvement of the NAD+ cellular content, an increase of PARylation of cardiac proteins and an improvement of left ventricular structure and function in a model of LMNA cardiomyopathy. Collectively, our results provide mechanistic and therapeutic insights into dilated cardiomyopathy caused by LMNA mutations.
Collapse
Affiliation(s)
- Nicolas Vignier
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Maria Chatzifrangkeskou
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Blanca Morales Rodriguez
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Mathias Mericskay
- INSERM UMR-S 1180 - LabEx LERMIT - DHU TORINO, Institut Paris-Saclay d'Innovation Therapeutique (IPSIT-US31-UMS3679), Faculty of Pharmacy, Univ Paris-Sud, Université Paris-Saclay, Chatenay-Malabry, France
| | - Nathalie Mougenot
- Sorbonne Université, UPMC Paris 06, INSERM UMS28 Phénotypage du petit animal, Faculté de Médecine Pierre et Marie Curie, Paris, France
| | - Karim Wahbi
- Cardiology Department, Cochin Hospital, Filière Neuromusculaire, Paris-Descartes University, Sorbonne Paris Cité University, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Gisèle Bonne
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| | - Antoine Muchir
- Sorbonne Université, UPMC Paris 06, INSERM UMRS974, Center of Research in Myology, Institut de Myologie, Paris, France
| |
Collapse
|