1
|
Florczyk-Soluch U, Polak K, Jelinkova S, Bronisz-Budzyńska I, Sabo R, Bolisetty S, Agarwal A, Werner E, Józkowicz A, Stępniewski J, Szade K, Dulak J. Targeted expression of heme oxygenase-1 in satellite cells improves skeletal muscle pathology in dystrophic mice. Skelet Muscle 2024; 14:13. [PMID: 38867250 PMCID: PMC11167827 DOI: 10.1186/s13395-024-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.
Collapse
MESH Headings
- Animals
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mice, Inbred mdx
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Male
- Mice, Inbred C57BL
- Physical Conditioning, Animal
- Membrane Proteins
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Sarka Jelinkova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Reece Sabo
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Laboratory of Stem Cells Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
2
|
Xiao Q, Sun CC, Tang CF. Heme oxygenase-1: A potential therapeutic target for improving skeletal muscle atrophy. Exp Gerontol 2023; 184:112335. [PMID: 37984695 DOI: 10.1016/j.exger.2023.112335] [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: 10/07/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Skeletal muscle atrophy is a common muscle disease that is directly caused by an imbalance in protein synthesis and degradation. At the histological level, it is mainly characterized by a reduction in muscle mass and fiber cross-sectional area (CSA). Patients with skeletal muscle atrophy present with reduced motor ability, easy fatigue, and poor life quality. Heme oxygenase-1 (HO-1) is an inducible enzyme that catalyzes the degradation of heme and has attracted much attention for its anti-oxidation effects. In addition, there is growing evidence that HO-1 plays an important role in anti-inflammatory, anti-apoptosis, pro-angiogenesis, and maintaining skeletal muscle homeostasis, making it a potential therapeutic target for improving skeletal muscle atrophy. Here, we review the pathogenesis of skeletal muscle atrophy, the biology of HO-1 and its regulation, and the biological function of HO-1 in skeletal muscle homeostasis, with a specific focus on the role of HO-1 in skeletal muscle atrophy, aiming to observe the therapeutic potential of HO-1 for skeletal muscle atrophy.
Collapse
Affiliation(s)
- Qin Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China; School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China
| | - Chen-Chen Sun
- School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China.
| |
Collapse
|
3
|
Myszka M, Mucha O, Podkalicka P, Waśniowska U, Dulak J, Łoboda A. Sodium hydrosulfide moderately alleviates the hallmark symptoms of Duchenne muscular dystrophy in mdx mice. Eur J Pharmacol 2023; 955:175928. [PMID: 37507045 DOI: 10.1016/j.ejphar.2023.175928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/22/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an incurable disease caused by mutations in the X-linked DMD gene that encodes a structural muscle protein, dystrophin. This, in turn, leads to progressive degeneration of the skeletal muscles and the heart. Hydrogen sulfide (H2S), the pleiotropic agent with antioxidant, anti-inflammatory, and pro-angiogenic activities, could be considered a promising therapeutic factor for DMD. In this work, we studied the effect of daily intraperitoneal administration of the H2S donor, sodium hydrosulfide (NaHS, 100 μmol/kg/day for 5 weeks) on skeletal muscle (gastrocnemius, diaphragm and tibialis anterior) pathology in dystrophin-deficient mdx mice, characterized by decreased expression of H2S-generating enzymes. NaHS reduced the level of muscle damage markers in plasma (creatine kinase, lactate dehydrogenase and osteopontin). It lowered oxidative stress by affecting the GSH/GSSG ratio, up-regulating the level of cytoprotective heme oxygenase-1 (HO-1) and down-regulating the NF-κB pathway. In the gastrocnemius muscle, it also increased angiogenic vascular endothelial growth factor (Vegf) and its receptor (Kdr) expression, accompanied by the elevated number of α-SMA/CD31/lectin-positive blood vessels. The expression of fibrotic regulators, like Tgfβ, Col1a1 and Fn1 was decreased by NaHS in the tibialis anterior, while the level of autophagy markers (AMPKα signalling and Atg genes), was mostly affected in the gastrocnemius. Histological and molecular analysis showed no effect of H2S donor on regeneration and the muscle fiber type composition. Overall, the H2S donor modified the gene expression and protein level of molecules associated with the pathophysiology of DMD, contributing to the regulation of oxidative stress, inflammation, autophagy, and angiogenesis.
Collapse
Affiliation(s)
- Małgorzata Myszka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Prof. St. Łojasiewicz 11, 30-348, Krakow, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Urszula Waśniowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland.
| |
Collapse
|
4
|
Effect of heme oxygenase-1 on the differentiation of human myoblasts and the regeneration of murine skeletal muscles after acute and chronic injury. Pharmacol Rep 2023; 75:397-410. [PMID: 36918494 DOI: 10.1007/s43440-023-00475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Impaired muscle regeneration is a hallmark of Duchenne muscular dystrophy (DMD), a neuromuscular disorder caused by mutations in the DMD gene encoding dystrophin. The lack of heme oxygenase-1 (HO-1, Hmox1), a known anti-inflammatory and cytoprotective enzyme, was shown to aggravate DMD pathology. METHODS We evaluated the role of HO-1 overexpression in human induced pluripotent stem cell (hiPSC)-derived skeletal muscle cells (hiPSC-SkM) in vitro and in the regeneration process in vivo in wild-type mice. Furthermore, the effect of cobalt protoporphyrin IX (CoPP), a pharmacological inducer of HO-1 expression, on regeneration markers during myogenic hiPSC differentiation and progression of the dystrophic phenotype was analysed in the mdx mouse DMD model. RESULTS HO-1 has an impact on hiPSC-SkM generation by decreasing cell fusion capacity and the expression of myogenic regulatory factors and muscle-specific microRNAs (myomiRs). Also, strong induction of HO-1 by CoPP totally abolished hiPSC-SkM differentiation. Injection of HO-1-overexpressing hiPSC-SkM into the cardiotoxin (CTX)-injured muscle of immunodeficient wild-type mice was associated with decreased expression of miR-206 and Myh3 and lower number of regenerating fibers, suggesting some advanced regeneration. However, the very potent induction of HO-1 by CoPP did not exert any protective effect on necrosis, leukocyte infiltration, fibrosis, myofiber regeneration biomarkers, and exercise capacity of mdx mice. CONCLUSIONS In summary, HO-1 inhibits the expression of differentiation markers in human iPSC-derived myoblasts. Although moderate overexpression of HO-1 in the injected myoblast was associated with partially advanced muscle regeneration, the high systemic induction of HO-1 did not improve muscle regeneration. The appropriate threshold of HO-1 expression must be established for the therapeutic effect of HO-1 on muscle regeneration.
Collapse
|
5
|
Hao Y, Xue T, Liu S, Geng S, Shi X, Qian P, He W, Zheng J, Li Y, Lou J, Shi T, Wang G, Wang X, Wang Y, Li Y, Song Y. Loss of CRY2 promotes regenerative myogenesis by enhancing PAX7 expression and satellite cell proliferation. MedComm (Beijing) 2023; 4:e202. [PMID: 36636367 PMCID: PMC9830134 DOI: 10.1002/mco2.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
The regenerative capacity of skeletal muscle is dependent on satellite cells. The circadian clock regulates the maintenance and function of satellite cells. Cryptochrome 2 (CRY2) is a critical component of the circadian clock, and its role in skeletal muscle regeneration remains controversial. Using the skeletal muscle lineage and satellite cell-specific CRY2 knockout mice (CRY2scko), we show that the deletion of CRY2 enhances muscle regeneration. Single myofiber analysis revealed that deletion of CRY2 stimulates the proliferation of myoblasts. The differentiation potential of myoblasts was enhanced by the loss of CRY2 evidenced by increased expression of myosin heavy chain (MyHC) and myotube formation in CRY2-/- cells versus CRY2+/+ cells. Immunostaining revealed that the number of mononucleated paired box protein 7 (PAX7+) cells associated with myotubes formed by CRY2-/- cells was increased compared with CRY2+/+ cells, suggesting that more reserve cells were produced in the absence of CRY2. Loss of CRY2 leads to the activation of the ERK1/2 signaling pathway and ETS1, which binds to the promoter of PAX7 to induce its transcription. CRY2 deficient myoblasts survived better in ischemic muscle. Therefore, CRY2 is essential in regulating skeletal muscle repair.
Collapse
Affiliation(s)
- Yingxue Hao
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ting Xue
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Song‐Bai Liu
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Sha Geng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xinghong Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Panting Qian
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Wei He
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jiqing Zheng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Yanfang Li
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jing Lou
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Tianze Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ge Wang
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xiaoxiao Wang
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Yanli Wang
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yangxin Li
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yao‐Hua Song
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| |
Collapse
|
6
|
Zheng T, Huang Z, Ling H, Li J, Cheng H, Chen D, Lu Q, Zhao J, Su W. The mechanism of the Nfe2l2/Hmox1 signaling pathway in ferroptosis regulation in acute compartment syndrome. J Biochem Mol Toxicol 2023; 37:e23228. [PMID: 36193742 PMCID: PMC10078270 DOI: 10.1002/jbt.23228] [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: 10/30/2021] [Revised: 07/30/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Acute compartment syndrome (ACS) is a life-threatening orthopedic emergency, which can even result in amputation. Ferroptosis is an iron-dependent form of nonapoptotic cell death. This study investigated the mechanism of ferroptosis in ACS, explored candidate markers, and determined effective treatments. This study identified pathways involved in the development of ACS through gene set enrichment analysis (GSEA), Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG), and GSEA of heme oxygenase 1 (Hmox1). Bioinformatics methods, combined with real-time quantitative polymerase chain reaction, western blot analysis, and iron staining, were applied to determine whether ferroptosis was involved in the progression of ACS and to explore the mechanism of nuclear factor erythroid-2-related factor 2 (Nfe2l2)/Hmox1 in ferroptosis regulation. Optimal drugs for the treatment of ACS were also investigated using Connectivity Map. The ferroptosis pathway was enriched in GSEA, KEGG of DEGs, and GSEA of Hmox1. After ACS, the reactive oxygen species content, tissue iron content, and oxidative stress level increased, whereas glutathione peroxidase 4 protein expression decreased. The skeletal muscle was swollen and necrotized; the number of mitochondrial cristae became fewer or even disappeared, and Nfe2l2/Hmox1 expression increased at the transcriptional and protein levels. Hmox1 was highly expressed in ACS, indicating that Hmox1 is a possible marker for ACS. we could predict 12 potential target drugs for the treatment of ACS. In conclusion, Hmox1 was a potential candidate marker for ACS diagnosis. Ferroptosis was involved in the progression of ACS. It was speculated that ferroptosis is inhibited by the Nfe2l2/Hmox1 signaling pathway.
Collapse
Affiliation(s)
- Tiejun Zheng
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Zhao Huang
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - He Ling
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Junfeng Li
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Cheng
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Dingquan Chen
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Qinzhen Lu
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Su
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
7
|
Chen Z, Lan H, Liao Z, Huang J, Jian X, Hu J, Liao H. Regulatory T cells-centered regulatory networks of skeletal muscle inflammation and regeneration. Cell Biosci 2022; 12:112. [PMID: 35869487 PMCID: PMC9308315 DOI: 10.1186/s13578-022-00847-x] [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: 09/27/2021] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
As the understanding of skeletal muscle inflammation is increasingly clarified, the role of Treg cells in the treatment of skeletal muscle diseases has attracted more attention in recent years. A consensus has been reached that the regulation of Treg cells is the key to completing the switch of inflammation and repair of skeletal muscle, whose presence directly determine the repairing quality of the injured skeletal muscle. However, the functioning process of Treg cells remains unreported, thereby making it necessary to summarize the current role of Treg cells in skeletal muscle. In this review, the characteristics, origins, and cellular kinetics of these Treg cells are firstly described; Then, the relationship between Treg cells and muscle satellite cells (MuSCs), conventional T cells (Tconv) is discussed (the former is involved in the entire repair and regeneration process, while the latter matters considerably in causing most skeletal muscle autoimmune diseases); Next, focus is placed on the control of Treg cells on the phenotypic switch of macrophages, which is the key to the switch of inflammation; Finally, factors regulating the functional process of Treg cells are analyzed, and a regulatory network centered on Treg cells is summarized. The present study summarizes the cell-mediated interactions in skeletal muscle repair over the past decade, and elucidates the central role of regulatory T cells in this process, so that other researchers can more quickly and comprehensively understand the development and direction of this very field. It is believed that the hereby proposed viewpoints and problems can provide fresh visions for the latecomers.
Collapse
|
8
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
9
|
Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Madej M, Józkowicz A, Łoboda A, Dulak J. NRF2 Regulates Viability, Proliferation, Resistance to Oxidative Stress, and Differentiation of Murine Myoblasts and Muscle Satellite Cells. Cells 2022; 11:cells11203321. [PMID: 36291188 PMCID: PMC9600498 DOI: 10.3390/cells11203321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Increased oxidative stress can slow down the regeneration of skeletal muscle and affect the activity of muscle satellite cells (mSCs). Therefore, we evaluated the role of the NRF2 transcription factor (encoded by the Nfe2l2 gene), the main regulator of the antioxidant response, in muscle cell biology. We used (i) an immortalized murine myoblast cell line (C2C12) with stable overexpression of NRF2 and (ii) primary mSCs isolated from wild-type and Nfe2l2 (transcriptionally)-deficient mice (Nfe2l2tKO). NRF2 promoted myoblast proliferation and viability under oxidative stress conditions and decreased the production of reactive oxygen species. Furthermore, NRF2 overexpression inhibited C2C12 cell differentiation by down-regulating the expression of myogenic regulatory factors (MRFs) and muscle-specific microRNAs. We also showed that NRF2 is indispensable for the viability of mSCs since the lack of its transcriptional activity caused high mortality of cells cultured in vitro under normoxic conditions. Concomitantly, Nfe2l2tKO mSCs grown and differentiated under hypoxic conditions were viable and much more differentiated compared to cells isolated from wild-type mice. Taken together, NRF2 significantly influences the properties of myoblasts and muscle satellite cells. This effect might be modulated by the muscle microenvironment.
Collapse
|
10
|
Leal EC, Carvalho E. Heme Oxygenase-1 as Therapeutic Target for Diabetic Foot Ulcers. Int J Mol Sci 2022; 23:ijms231912043. [PMID: 36233341 PMCID: PMC9569859 DOI: 10.3390/ijms231912043] [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: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022] Open
Abstract
A diabetic foot ulcer (DFU) is one of the major complications of diabetes. Wound healing under diabetic conditions is often impaired. This is in part due to the excessive oxidative stress, prolonged inflammation, immune cell dysfunction, delayed re-epithelialization, and decreased angiogenesis present at the wound site. As a result of these multifactorial impaired healing pathways, it has been difficult to develop effective therapeutic strategies for DFU. Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme degradation generating carbon monoxide (CO), biliverdin (BV) which is converted into bilirubin (BR), and iron. HO-1 is a potent antioxidant. It can act as an anti-inflammatory, proliferative, angiogenic and cytoprotective enzyme. Due to its biological functions, HO-1 plays a very important role in wound healing, in part mediated through the biologically active end products generated by its enzymatic activity, particularly CO, BV, and BR. Therapeutic strategies involving the activation of HO-1, or the topical application of its biologically active end products are important in diabetic wound healing. Therefore, HO-1 is an attractive therapeutic target for DFU treatment. This review will provide an overview and discussion of the importance of HO-1 as a therapeutic target for diabetic wound healing.
Collapse
Affiliation(s)
- Ermelindo Carreira Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
- Correspondence: (E.C.L.); (E.C.); Tel.: +351-239-820-190 (E.C.L. & E.C.)
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
- Correspondence: (E.C.L.); (E.C.); Tel.: +351-239-820-190 (E.C.L. & E.C.)
| |
Collapse
|
11
|
Olie CS, van Zeijl R, El Abdellaoui S, Kolk A, Overbeek C, Nelissen RGHH, Heijs B, Raz V. The metabolic landscape in chronic rotator cuff tear reveals tissue-region-specific signatures. J Cachexia Sarcopenia Muscle 2022; 13:532-543. [PMID: 34866353 PMCID: PMC8818701 DOI: 10.1002/jcsm.12873] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Degeneration of shoulder muscle tissues often result in tearing, causing pain, disability and loss of independence. Differential muscle involvement patterns have been reported in tears of shoulder muscles, yet the molecules involved in this pathology are poorly understood. The spatial distribution of biomolecules across the affected tissue can be accurately obtained with matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). The goal of this pilot study was to decipher the metabolic landscape across shoulder muscle tissues and to identify signatures of degenerated muscles in chronic conditions. METHODS Paired biopsies of two rotator cuff muscles, torn infraspinatus and intact teres minor, together with an intact shoulder muscle, the deltoid, were collected during an open tendon transfer surgery. Five patients, average age 65.2 ± 3.8 years, were selected for spatial metabolic profiling using high-spatial resolution (MALDI-TOF) and high-mass resolution (MALDI-FTICR) MSI in negative or positive ion mode. Metabolic signatures were identified using data-driven analysis. Verifications of spatial localization for selected metabolic signatures were carried out using antibody immunohistology. RESULTS Data-driven analysis revealed major metabolic differences between intact and degenerated regions across all muscles. The area of degenerated regions, encompassed of fat, inflammation and fibrosis, significantly increased in both rotator cuff muscles, teres minor (27.9%) and infraspinatus (22.8%), compared with the deltoid (8.7%). The intact regions were characterized by 49 features, among which lipids were recognized. Several of the identified lipids were specifically enriched in certain myofiber types. Degenerated regions were specifically marked by the presence of 37 features. Heme was the most abundant metabolite in degenerated regions, whereas Heme oxygenase-1 (HO-1), which catabolizes heme, was found in intact regions. Higher HO-1 levels correlated with lower heme accumulation. CONCLUSIONS Degenerated regions are distinguished from intact regions by their metabolome profile. A muscle-specific metabolome profile was not identified. The area of tissue degeneration significantly differs between the three examined muscles. Higher HO-1 levels in intact regions concurred with lower heme levels in degenerated regions. Moreover, HO-1 levels discriminated between dysfunctional and functional rotator cuff muscles. Additionally, the enrichment of specific lipids in certain myofiber types suggests that lipid metabolism differs between myofiber types. The signature metabolites can open options to develop personalized treatments for chronic shoulder muscles degeneration.
Collapse
Affiliation(s)
| | - René van Zeijl
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Salma El Abdellaoui
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjen Kolk
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Celeste Overbeek
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Vered Raz
- Human Genetics Department, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Kaziród K, Myszka M, Dulak J, Łoboda A. Hydrogen sulfide as a therapeutic option for the treatment of Duchenne muscular dystrophy and other muscle-related diseases. Cell Mol Life Sci 2022; 79:608. [PMID: 36441348 PMCID: PMC9705465 DOI: 10.1007/s00018-022-04636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Hydrogen sulfide (H2S) has been known for years as a poisoning gas and until recently evoked mostly negative associations. However, the discovery of its gasotransmitter functions suggested its contribution to various physiological and pathological processes. Although H2S has been found to exert cytoprotective effects through modulation of antioxidant, anti-inflammatory, anti-apoptotic, and pro-angiogenic responses in a variety of conditions, its role in the pathophysiology of skeletal muscles has not been broadly elucidated so far. The classical example of muscle-related disorders is Duchenne muscular dystrophy (DMD), the most common and severe type of muscular dystrophy. Mutations in the DMD gene that encodes dystrophin, a cytoskeletal protein that protects muscle fibers from contraction-induced damage, lead to prominent dysfunctions in the structure and functions of the skeletal muscle. However, the main cause of death is associated with cardiorespiratory failure, and DMD remains an incurable disease. Taking into account a wide range of physiological functions of H2S and recent literature data on its possible protective role in DMD, we focused on the description of the 'old' and 'new' functions of H2S, especially in muscle pathophysiology. Although the number of studies showing its essential regulatory action in dystrophic muscles is still limited, we propose that H2S-based therapy has the potential to attenuate the progression of DMD and other muscle-related disorders.
Collapse
Affiliation(s)
- Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Małgorzata Myszka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
13
|
Mucha O, Podkalicka P, Kaziród K, Samborowska E, Dulak J, Łoboda A. Simvastatin does not alleviate muscle pathology in a mouse model of Duchenne muscular dystrophy. Skelet Muscle 2021; 11:21. [PMID: 34479633 PMCID: PMC8414747 DOI: 10.1186/s13395-021-00276-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an incurable disease, caused by the mutations in the DMD gene, encoding dystrophin, an actin-binding cytoskeletal protein. Lack of functional dystrophin results in muscle weakness, degeneration, and as an outcome cardiac and respiratory failure. As there is still no cure for affected individuals, the pharmacological compounds with the potential to treat or at least attenuate the symptoms of the disease are under constant evaluation. The pleiotropic agents, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, known as statins, have been suggested to exert beneficial effects in the mouse model of DMD. On the other hand, they were also reported to induce skeletal-muscle myopathy. Therefore, we decided to verify the hypothesis that simvastatin may be considered a potential therapeutic agent in DMD. Methods Several methods including functional assessment of muscle function via grip strength measurement, treadmill test, and single-muscle force estimation, enzymatic assays, histological analysis of muscle damage, gene expression evaluation, and immunofluorescence staining were conducted to study simvastatin-related alterations in the mdx mouse model of DMD. Results In our study, simvastatin treatment of mdx mice did not result in improved running performance, grip strength, or specific force of the single muscle. Creatine kinase and lactate dehydrogenase activity, markers of muscle injury, were also unaffected by simvastatin delivery in mdx mice. Furthermore, no significant changes in inflammation, fibrosis, and angiogenesis were noted. Despite the decreased percentage of centrally nucleated myofibers in gastrocnemius muscle after simvastatin delivery, no changes were noticed in other regeneration-related parameters. Of note, even an increased rate of necrosis was found in simvastatin-treated mdx mice. Conclusion In conclusion, our study revealed that simvastatin does not ameliorate DMD pathology. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00276-3.
Collapse
Affiliation(s)
- Olga Mucha
- 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
| | - Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Emilia Samborowska
- Mass Spectrometry Lab, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warszawa, Poland
| | - Józef Dulak
- 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.
| |
Collapse
|
14
|
Liang Y, Yu H, Ke X, Eyles D, Sun R, Wang Z, Huang S, Lin L, McGrath JJ, Lu J, Guo X, Yao P. Vitamin D deficiency worsens maternal diabetes induced neurodevelopmental disorder by potentiating hyperglycemia-mediated epigenetic changes. Ann N Y Acad Sci 2021; 1491:74-88. [PMID: 33305416 DOI: 10.1111/nyas.14535] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/15/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022]
Abstract
Many studies have shown that vitamin D (VD) deficiency may be a risk factor for neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia, although causative mechanisms remain unknown. In this study, we investigated the potential role and effect of VD on maternal diabetes induced autism-related phenotypes. The in vitro study found that enhancing genomic VD signaling by overexpressing the VD receptor (VDR) in human neural progenitor cells ACS-5003 protects against hyperglycemia-induced oxidative stress and inflammation by activating Nrf2 and its target genes, including SOD2 and HMOX1, and accordingly, VDR gene knockdown worsens the problem. In the two in vivo models we explored, maternal diabetes was used to establish an animal model of relevance to ASD, and mice lacking 25-hydroxyvitamin D 1-alpha-hydroxylase (the rate-limiting enzyme in the synthesis of 1,25(OH)2D3) were used to develop a model of VD deficiency (VDD). We show that although prenatal VDD itself does not produce ASD-relevant phenotypes, it significantly potentiates maternal diabetes induced epigenetic modifications and autism-related phenotypes. Postnatal manipulation of VD has no effect on maternal diabetes induced autism-related phenotypes. We conclude that VDD potentiates maternal diabetes induced autism-related phenotypes in offspring by epigenetic mechanisms. This study adds to other preclinical studies linking prenatal VDD with a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Hong Yu
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Xiaoyin Ke
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Darryl Eyles
- Queensland Centre for Mental Health Research, the Park Centre for Mental Health, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland, Australia
| | - Ruoyu Sun
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Saijun Huang
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Ling Lin
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - John J McGrath
- Queensland Centre for Mental Health Research, the Park Centre for Mental Health, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland, Australia
- National Centre for Register-based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
| | - Jianping Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Xiaoling Guo
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Paul Yao
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| |
Collapse
|
15
|
Age-Dependent Dysregulation of Muscle Vasculature and Blood Flow Recovery after Hindlimb Ischemia in the mdx Model of Duchenne Muscular Dystrophy. Biomedicines 2021; 9:biomedicines9050481. [PMID: 33925757 PMCID: PMC8145677 DOI: 10.3390/biomedicines9050481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by a lack of functional dystrophin, is characterized by progressive muscle degeneration. Interestingly, dystrophin is also expressed in endothelial cells (ECs), and insufficient angiogenesis has already been hypothesized to contribute to DMD pathology, however, its status in mdx mice, a model of DMD, is still not fully clear. Our study aimed to reveal angiogenesis-related alterations in skeletal muscles of mdx mice compared to wild-type (WT) counterparts. By investigating 6- and 12-week-old mice, we sought to verify if those changes are age-dependent. We utilized a broad spectrum of methods ranging from gene expression analysis, flow cytometry, and immunofluorescence imaging to determine the level of angiogenic markers and to assess muscle blood vessel abundance. Finally, we implemented the hindlimb ischemia (HLI) model, more biologically relevant in the context of functional studies evaluating angiogenesis/arteriogenesis processes. We demonstrated that both 6- and 12-week-old dystrophic mice exhibited dysregulation of several angiogenic factors, including decreased vascular endothelial growth factor A (VEGF) in different muscle types. Nonetheless, in younger, 6-week-old mdx animals, neither the abundance of CD31+α-SMA+ double-positive blood vessels nor basal blood flow and its restoration after HLI was affected. In 12-week-old mdx mice, although a higher number of CD31+α-SMA+ double-positive blood vessels and an increased percentage of skeletal muscle ECs were found, the abundance of pericytes was diminished, and blood flow was reduced. Moreover, impeded perfusion recovery after HLI associated with a blunted inflammatory and regenerative response was evident in 12-week-old dystrophic mice. Hence, our results reinforce the hypothesis of age-dependent angiogenic dysfunction in dystrophic mice. In conclusion, we suggest that older mdx mice constitute an appropriate model for preclinical studies evaluating the effectiveness of vascular-based therapies aimed at the restoration of functional angiogenesis to mitigate DMD severity.
Collapse
|
16
|
TAKAI Y, WATANABE T, SANO T. Elevated level of microRNA-210 at the initiation of muscular regeneration in acetic acid-induced non-ischemic skeletal muscular injury in mice. J Toxicol Pathol 2021; 35:183-192. [PMID: 35516838 PMCID: PMC9018401 DOI: 10.1293/tox.2021-0061] [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: 09/15/2021] [Accepted: 11/29/2021] [Indexed: 11/29/2022] Open
Abstract
The alteration in microRNA-210 level, a hypoxia-inducible microRNA, is not well known in
non-ischemic tissue injury. In this study, we characterized the histopathological time
course of acetic acid-induced skeletal muscle injury as a non-ischemic tissue injury model
and investigated the expression of microRNA-210, hypoxia-inducible factor 1α, and growth
factors using quantitative polymerase chain reaction analysis. After a single
intramuscular dose of 3% (v/v) acetic acid to C57BL/6J mice, focal coagulative necrosis of
muscle fibers was noted from 3 h after dosing and infiltration of F4/80 and Galectin-3
positive M2 macrophage was noted at 1 d after dosing. Muscular regeneration was initiated
from 3 d, when M2 macrophage infiltration was most prominent, till 14 d after dosing.
Hif1α and Hgf expression increased from 3 h onwards,
and microRNA-210 level increased after 3 d after the treatment. However, no clear
elevation in the levels of Igf1 or Vegf was observed.
The infiltrative macrophages and regenerative muscle fibers were positive for
hypoxia-inducible factor 1α, microRNA-210, and hepatocyte growth factor as assessed by
immunohistochemistry or in situ hybridization. In this study, dominant
infiltration of M2 macrophages at muscular necrosis and subsequent regeneration after a
single intramuscular injection of acetic acid in mice were observed. The increase in hif1α
level was observed just after the muscular injury in this non-ischemic tissue injury
model, and the elevation in microRNA-210 level was noted at the initiation of tissue
regeneration, indicating its effects on tissue protection and repair.
Collapse
Affiliation(s)
- Yuichi TAKAI
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2 Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi WATANABE
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2 Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomoya SANO
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2 Chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
17
|
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: 1.8] [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
|
18
|
Łoboda A, Dulak J. Muscle and cardiac therapeutic strategies for Duchenne muscular dystrophy: past, present, and future. Pharmacol Rep 2020; 72:1227-1263. [PMID: 32691346 PMCID: PMC7550322 DOI: 10.1007/s43440-020-00134-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular childhood disorder that causes progressive muscle weakness and degeneration and results in functional decline, loss of ambulation and early death of young men due to cardiac or respiratory failure. Although the major cause of the disease has been known for many years-namely mutation in the DMD gene encoding dystrophin, one of the largest human genes-DMD is still incurable, and its treatment is challenging. METHODS A comprehensive and systematic review of literature on the gene, cell, and pharmacological experimental therapies aimed at restoring functional dystrophin or to counteract the associated processes contributing to disease progression like inflammation, fibrosis, calcium signaling or angiogenesis was carried out. RESULTS Although some therapies lead to satisfying effects in skeletal muscle, they are highly ineffective in the heart; therefore, targeting defective cardiac and respiratory systems is vital in DMD patients. Unfortunately, most of the pharmacological compounds treat only the symptoms of the disease. Some drugs addressing the underlying cause, like eteplirsen, golodirsen, and ataluren, have recently been conditionally approved; however, they can correct only specific mutations in the DMD gene and are therefore suitable for small sub-populations of affected individuals. CONCLUSION In this review, we summarize the possible therapeutic options and describe the current status of various, still imperfect, strategies used for attenuating the disease progression.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
19
|
Tift MS, Alves de Souza RW, Weber J, Heinrich EC, Villafuerte FC, Malhotra A, Otterbein LE, Simonson TS. Adaptive Potential of the Heme Oxygenase/Carbon Monoxide Pathway During Hypoxia. Front Physiol 2020; 11:886. [PMID: 32792988 PMCID: PMC7387684 DOI: 10.3389/fphys.2020.00886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Heme oxygenase (HO) enzymes catalyze heme into biliverdin, releasing carbon monoxide (CO) and iron into circulation. These byproducts of heme degradation can have potent cytoprotective effects in the face of stressors such as hypoxia and ischemia-reperfusion events. The potential for exogenous use of CO as a therapeutic agent has received increasing attention throughout the past few decades. Further, HO and CO are noted as putatively adaptive in diving mammals and certain high-altitude human populations that are frequently exposed to hypoxia and/or ischemia-reperfusion events, suggesting that HO and endogenous CO afford an evolutionary advantage for hypoxia tolerance and are critical in cell survival and injury avoidance. Our goal is to describe the importance of examining HO and CO in several systems, the physiological links, and the genetic factors that underlie variation in the HO/CO pathway. Finally, we emphasize the ways in which evolutionary perspectives may enhance our understanding of the HO/CO pathway in the context of diverse clinical settings.
Collapse
Affiliation(s)
- Michael S. Tift
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Rodrigo W. Alves de Souza
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Janick Weber
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Erica C. Heinrich
- Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, United States
| | - Francisco C. Villafuerte
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, School of Medicine, San Diego, CA, United States
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, School of Medicine, San Diego, CA, United States
| |
Collapse
|
20
|
Podkalicka P, Mucha O, Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Cetnarowska A, Głowniak-Kwitek U, Bukowska-Strakova K, Cieśla M, Kulecka M, Ostrowski J, Mikuła M, Potulska-Chromik A, Kostera-Pruszczyk A, Józkowicz A, Łoboda A, Dulak J. Lack of miR-378 attenuates muscular dystrophy in mdx mice. JCI Insight 2020; 5:135576. [PMID: 32493839 PMCID: PMC7308053 DOI: 10.1172/jci.insight.135576] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/29/2020] [Indexed: 01/09/2023] Open
Abstract
The severity of Duchenne muscular dystrophy (DMD), an incurable disease caused by the lack of dystrophin, might be modulated by different factors, including miRNAs. Among them, miR-378 is considered of high importance for muscle biology, but intriguingly, its role in DMD and its murine model (mdx mice) has not been thoroughly addressed so far. Here, we demonstrate that dystrophic mice additionally globally lacking miR-378 (double-KO [dKO] animals) exhibited better physical performance and improved absolute muscle force compared with mdx mice. Accordingly, markers of muscle damage in serum were significantly decreased in dKO mice, accompanied by diminished inflammation, fibrosis, and reduced abundance of regenerating fibers within muscles. The lack of miR-378 also normalized the aggravated fusion of dystrophin-deficient muscle satellite cells (mSCs). RNA sequencing of gastrocnemius muscle transcriptome revealed fibroblast growth factor 1 (Fgf1) as one of the most significantly downregulated genes in mice devoid of miR-378, indicating FGF1 as one of the mediators of changes driven by the lack of miR-378. In conclusion, we suggest that targeting miR-378 has the potential to ameliorate DMD pathology.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | | | - Anna Cetnarowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Urszula Głowniak-Kwitek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and.,Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Medical College, Jagiellonian University, Krakow, Poland
| | - Maciej Cieśla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland.,Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Michał Mikuła
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | | | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| |
Collapse
|
21
|
Peterson SJ, Dave N, Kothari J. The Effects of Heme Oxygenase Upregulation on Obesity and the Metabolic Syndrome. Antioxid Redox Signal 2020; 32:1061-1070. [PMID: 31880952 DOI: 10.1089/ars.2019.7954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Obesity is a chronic condition that is characterized by inflammation and oxidative stress with consequent cardiovascular complications of hypertension, dyslipidemia, and vascular dysfunction. Obesity-induced metabolic syndrome remains an epidemic of global proportions. Recent Advances: Gene targeting of the endothelium with a retrovirus using an endothelium-specific promoter vascular endothelium cadherin (VECAD)-HO-1 offers a potential long-term solution to adiposity by targeting the endothelium. This has resulted in improvements of both vascular function and adiposity attenuation. Critical Issues: Heme oxygenase plays an ever-increasing role in the understanding of human biology in the complex conditions of obesity and the metabolic syndrome. The heme oxygenase 1 (HO-1) system creates biliverdin/bilirubin, which functions as an antioxidant, and carbon monoxide, which has antiapoptotic properties. Future Directions: Upregulation of HO-1 has been shown to improve adiposity as well as vascular function in both animal and human studies.
Collapse
Affiliation(s)
- Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, New York.,New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, New York
| | - Niel Dave
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, New York
| | - Janish Kothari
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, New York
| |
Collapse
|
22
|
Pratt R, Lakhani HV, Zehra M, Desauguste R, Pillai SS, Sodhi K. Mechanistic Insight of Na/K-ATPase Signaling and HO-1 into Models of Obesity and Nonalcoholic Steatohepatitis. Int J Mol Sci 2019; 21:ijms21010087. [PMID: 31877680 PMCID: PMC6982200 DOI: 10.3390/ijms21010087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a multifaceted pathophysiological condition that has been associated with lipid accumulation, adipocyte dysfunction, impaired mitochondrial biogenesis and an altered metabolic profile. Redox imbalance and excessive release of inflammatory mediators have been intricately linked in obesity-associated phenotypes. Hence, understanding the mechanisms of redox signaling pathways and molecular targets exacerbating oxidative stress is crucial in improving health outcomes. The activation of Na/K-ATPase/Src signaling, and its downstream pathways, by reactive oxygen species (ROS) has been recently implicated in obesity and subsequent nonalcoholic steatohepatitis (NASH), which causes further production of ROS creating an oxidant amplification loop. Apart from that, numerous studies have also characterized antioxidant properties of heme oxygenase 1 (HO-1), which is suppressed in an obese state. The induction of HO-1 restores cellular redox processes, which contributes to inhibition of the toxic milieu. The novelty of these independent mechanisms presents a unique opportunity to unravel their potential as molecular targets for redox regulation in obesity and NASH. The attenuation of oxidative stress, by understanding the underlying molecular mechanisms and associated mediators, with a targeted treatment modality may provide for improved therapeutic options to combat clinical disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Komal Sodhi
- Correspondence: ; Tel.: +1-(304)-691-1704; Fax: +1-(914)-347-4956
| |
Collapse
|
23
|
Li M, Yu H, Pan H, Zhou X, Ruan Q, Kong D, Chu Z, Li H, Huang J, Huang X, Chau A, Xie W, Ding Y, Yao P. Nrf2 Suppression Delays Diabetic Wound Healing Through Sustained Oxidative Stress and Inflammation. Front Pharmacol 2019; 10:1099. [PMID: 31616304 PMCID: PMC6763603 DOI: 10.3389/fphar.2019.01099] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
Impaired wound healing is one of the major complications of diabetes, involving prolonged inflammation, delayed re-epithelialization, and consistent oxidative stress. The detailed mechanism remains unclear, and there is currently no effective treatment for diabetic wound healing. In this study, we aim to investigate the potential role and effect of nuclear factor erythroid-2–related factor-2 (Nrf2) activation on diabetic wound healing. In vitro experiments in rat macrophages showed that hyperglycemia treatment suppresses Nrf2 activation, resulting in oxidative stress with decreased expression of antioxidant genes, including NAD(P)H:quinone oxidoreductase 1 and heme oxygenase 1, together with increased secretion of proinflammatory cytokines, including interleukin 1β (IL1β), IL6, and monocyte chemoattractant protein-1. Both Nrf2 overexpression and Nrf2 activator dimethyl fumarate (DMF) treatment significantly ameliorated oxidative stress and inflammation. On the other hand, both Nrf2 knockdown or Nrf2 inhibitor ML385 mimicked the effect of diabetes. Further in vivo experiments in rats showed that DMF treatment significantly accelerated wound healing in streptozocin-induced diabetic rats with increased expression of antioxidant enzymes and decreased secretion of proinflammatory cytokines, while Nrf2 inhibitor ML385 mimicked the effect of diabetes. We conclude that Nrf2 activation accelerates impaired wound healing by ameliorating diabetes-mediated oxidative stress and inflammation. This provides a new clinical treatment strategy for diabetic wound healing using Nrf2 activator DMF.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Haibing Yu
- School of Public Health, Guangdong Medical College, Dongguan, China.,Key Lab of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haiyan Pan
- School of Public Health, Guangdong Medical College, Dongguan, China
| | - Xueqing Zhou
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Qiongfang Ruan
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Danli Kong
- School of Public Health, Guangdong Medical College, Dongguan, China
| | - Zhigang Chu
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Huawen Li
- School of Public Health, Guangdong Medical College, Dongguan, China
| | - Jingwen Huang
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Xiaodong Huang
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Angel Chau
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Weiguo Xie
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical College, Dongguan, China
| | - Paul Yao
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan Third Hospital, Wuhan, China.,School of Public Health, Guangdong Medical College, Dongguan, China
| |
Collapse
|
24
|
Bronisz-Budzyńska I, Chwalenia K, Mucha O, Podkalicka P, Karolina-Bukowska-Strakova, Józkowicz A, Łoboda A, Kozakowska M, Dulak J. miR-146a deficiency does not aggravate muscular dystrophy in mdx mice. Skelet Muscle 2019; 9:22. [PMID: 31412923 PMCID: PMC6693262 DOI: 10.1186/s13395-019-0207-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease evoked by a mutation in the dystrophin gene. It is associated with progressive muscle degeneration and increased inflammation. Up to this date, mainly anti-inflammatory treatment is available for patients suffering from DMD. miR-146a is known to diminish inflammation and fibrosis in different tissues by downregulating the expression of proinflammatory cytokines. However, its role in DMD has not been studied so far. In our work, we have generated mice globally lacking both dystrophin and miR-146a (miR-146a−/−mdx) and examined them together with wild-type, single miR-146a knockout and dystrophic (mdx—lacking dystrophin) mice in a variety of aspects associated with DMD pathophysiology (muscle degeneration, inflammatory reaction, muscle satellite cells, muscle regeneration, and fibrosis). We have shown that miR-146a level is increased in dystrophic muscles in comparison to wild-type mice. Its deficiency augments the expression of proinflammatory cytokines (IL-1β, CCL2, TNFα). However, muscle degeneration was not significantly worsened in mdx mice lacking miR-146a up to 24 weeks of age, although some aggravation of muscle damage and inflammation was evident in 12-week-old animals, though no effect of miR-146a deficiency was visible on quantity, proliferation, and in vitro differentiation of muscle satellite cells isolated from miR-146a−/−mdx mice vs. mdx. Similarly, muscle regeneration and collagen deposition were not changed by miR-146a deficiency. Nevertheless, the lack of miR-146a is associated with decreased Vegfa and increased Tgfb1. Overall, the lack of miR-146a did not aggravate significantly the dystrophic conditions in mdx mice, but its effect on DMD in more severe conditions warrants further investigation.
Collapse
Affiliation(s)
- Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Katarzyna Chwalenia
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Karolina-Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.,Department of Clinical Immunology and Transplantology, Institute of Paediatrics, Medical College, Jagiellonian University, Wielicka 265, 30-663, Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| |
Collapse
|
25
|
Patsalos A, Tzerpos P, Halasz L, Nagy G, Pap A, Giannakis N, Lyroni K, Koliaraki V, Pintye E, Dezso B, Kollias G, Spilianakis CG, Nagy L. The BACH1-HMOX1 Regulatory Axis Is Indispensable for Proper Macrophage Subtype Specification and Skeletal Muscle Regeneration. THE JOURNAL OF IMMUNOLOGY 2019; 203:1532-1547. [PMID: 31405954 DOI: 10.4049/jimmunol.1900553] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022]
Abstract
The infiltration and subsequent in situ subtype specification of monocytes to effector/inflammatory and repair macrophages is indispensable for tissue repair upon acute sterile injury. However, the chromatin-level mediators and regulatory events controlling this highly dynamic macrophage phenotype switch are not known. In this study, we used a murine acute muscle injury model to assess global chromatin accessibility and gene expression dynamics in infiltrating macrophages during sterile physiological inflammation and tissue regeneration. We identified a heme-binding transcriptional repressor, BACH1, as a novel regulator of this process. Bach1 knockout mice displayed impaired muscle regeneration, altered dynamics of the macrophage phenotype transition, and transcriptional deregulation of key inflammatory and repair-related genes. We also found that BACH1 directly binds to and regulates distal regulatory elements of these genes, suggesting a novel role for BACH1 in controlling a broad spectrum of the repair response genes in macrophages upon injury. Inactivation of heme oxygenase-1 (Hmox1), one of the most stringently deregulated genes in the Bach1 knockout in macrophages, impairs muscle regeneration by changing the dynamics of the macrophage phenotype switch. Collectively, our data suggest the existence of a heme-BACH1--HMOX1 regulatory axis, that controls the phenotype and function of the infiltrating myeloid cells upon tissue damage, shaping the overall tissue repair kinetics.
Collapse
Affiliation(s)
- Andreas Patsalos
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary.,Department of Biology, University of Crete, Heraklion, GR-70013 Greece
| | - Laszlo Halasz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Nikolas Giannakis
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Konstantina Lyroni
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, GR-71003 Greece
| | - Vasiliki Koliaraki
- Biomedical Sciences Research Center "Alexander Fleming," Vari, GR-16672 Greece
| | - Eva Pintye
- Department of Radiotherapy, University of Debrecen, Debrecen, HU-4032 Hungary.,Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701.,Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701
| | - Balazs Dezso
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701
| | - George Kollias
- Biomedical Sciences Research Center "Alexander Fleming," Vari, GR-16672 Greece.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| | - Charalampos G Spilianakis
- Department of Biology, University of Crete, Heraklion, GR-70013 Greece.,Department of Biology, University of Crete, Heraklion, GR-70013 Greece.,Department of Biology, University of Crete, Heraklion, GR-70013 Greece
| | - Laszlo Nagy
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701; .,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032 Hungary
| |
Collapse
|
26
|
LoPresti ST, Popovic B, Kulkarni M, Skillen CD, Brown BN. Free radical-decellularized tissue promotes enhanced antioxidant and anti-inflammatory macrophage response. Biomaterials 2019; 222:119376. [PMID: 31445321 DOI: 10.1016/j.biomaterials.2019.119376] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 07/15/2019] [Accepted: 07/21/2019] [Indexed: 12/19/2022]
Abstract
Oxidative stress leads to the progression of many diseases including chronic wounds, atherosclerosis, stroke and cancer. The modification of biomolecules with reactive nitrogen or oxygen species has been shown to trigger oxidative stress pathways that are beneficial for healing. Extracellular matrix scaffolds have been used successfully in reconstructive applications due to the beneficial host response they induce. To tailor extracellular matrix scaffolds to enhance antioxidant response, ECM were prepared using reactive nitrogen or oxygen species. These scaffolds were shown to be effectively decellularized and possess oxidative or nitroxidative protein modifications. Macrophage responses in vitro and in an in vivo muscle injury model were shown to have enhanced antioxidant phenotypes without impairment of long-term remodeling. These observations suggest that ECM decellularized with reactive oxygen or nitrogen species could provide better outcomes for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- S T LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, United States; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, United States
| | - B Popovic
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, United States
| | - M Kulkarni
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, United States
| | - C D Skillen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, United States
| | - B N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, United States; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, United States; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, United States.
| |
Collapse
|
27
|
Podkalicka P, Mucha O, Dulak J, Loboda A. Targeting angiogenesis in Duchenne muscular dystrophy. Cell Mol Life Sci 2019; 76:1507-1528. [PMID: 30770952 PMCID: PMC6439152 DOI: 10.1007/s00018-019-03006-7] [Citation(s) in RCA: 36] [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: 11/04/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) represents one of the most devastating types of muscular dystrophies which affect boys already at early childhood. Despite the fact that the primary cause of the disease, namely the lack of functional dystrophin is known already for more than 30 years, DMD still remains an incurable disease. Thus, an enormous effort has been made during recent years to reveal novel mechanisms that could provide therapeutic targets for DMD, especially because glucocorticoids treatment acts mostly symptomatic and exerts many side effects, whereas the effectiveness of genetic approaches aiming at the restoration of functional dystrophin is under the constant debate. Taking into account that dystrophin expression is not restricted to muscle cells, but is present also in, e.g., endothelial cells, alterations in angiogenesis process have been proposed to have a significant impact on DMD progression. Indeed, already before the discovery of dystrophin, several abnormalities in blood vessels structure and function have been revealed, suggesting that targeting angiogenesis could be beneficial in DMD. In this review, we will summarize current knowledge about the angiogenesis status both in animal models of DMD as well as in DMD patients, focusing on different organs as well as age- and sex-dependent effects. Moreover, we will critically discuss some approaches such as modulation of vascular endothelial growth factor or nitric oxide related pathways, to enhance angiogenesis and attenuate the dystrophic phenotype. Additionally, we will suggest the potential role of other mediators, such as heme oxygenase-1 or statins in those processes.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
28
|
Pietraszek-Gremplewicz K, Kozakowska M, Bronisz-Budzynska I, Ciesla M, Mucha O, Podkalicka P, Madej M, Glowniak U, Szade K, Stepniewski J, Jez M, Andrysiak K, Bukowska-Strakova K, Kaminska A, Kostera-Pruszczyk A, Jozkowicz A, Loboda A, Dulak J. Heme Oxygenase-1 Influences Satellite Cells and Progression of Duchenne Muscular Dystrophy in Mice. Antioxid Redox Signal 2018; 29:128-148. [PMID: 29669436 DOI: 10.1089/ars.2017.7435] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Muscle damage in Duchenne muscular dystrophy (DMD) caused by the lack of dystrophin is strongly linked to inflammation. Heme oxygenase-1 (HO-1; Hmox1) is an anti-inflammatory and cytoprotective enzyme affecting myoblast differentiation by inhibiting myomiRs. The role of HO-1 has not been so far well addressed in DMD. RESULTS In dystrophin-deficient mdx mice, expression of Hmox1 in limb skeletal muscles and diaphragm is higher than in wild-type animals, being consistently elevated from 8 up to 52 weeks, both in myofibers and inflammatory leukocytes. Accordingly, HO-1 expression is induced in muscles of DMD patients. Pharmacological inhibition of HO-1 activity or genetic ablation of Hmox1 aggravates muscle damage and inflammation in mdx mice. Double knockout animals (Hmox1-/-mdx) demonstrate impaired exercise capacity in comparison with mdx mice. Interestingly, in contrast to the effect observed in muscle fibers, in dystrophin-deficient muscle satellite cells (SCs) expression of Hmox1 is decreased, while MyoD, myogenin, and miR-206 are upregulated compared with wild-type counterparts. Mdx SCs demonstrate disturbed and enhanced differentiation, which is further intensified by Hmox1 deficiency. RNA sequencing revealed downregulation of Atf3, MafK, Foxo1, and Klf2 transcription factors, known to activate Hmox1 expression, as well as attenuation of nitric oxide-mediated cGMP-dependent signaling in mdx SCs. Accordingly, treatment with NO-donor induces Hmox1 expression and inhibits differentiation. Finally, differentiation of mdx SCs was normalized by CO, a product of HO-1 activity. Innovation and Conclusions: HO-1 is induced in DMD, and HO-1 inhibition aggravates DMD pathology. Therefore, HO-1 can be considered a therapeutic target to alleviate this disease. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Katarzyna Pietraszek-Gremplewicz
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Magdalena Kozakowska
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Iwona Bronisz-Budzynska
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Maciej Ciesla
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Olga Mucha
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Paulina Podkalicka
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Magdalena Madej
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Urszula Glowniak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Krzysztof Szade
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Jacek Stepniewski
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Mateusz Jez
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Kalina Andrysiak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Karolina Bukowska-Strakova
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland .,2 Department of Clinical Immunology and Transplantology, Institute of Paediatrics, Medical College, Jagiellonian University , Krakow, Poland
| | - Anna Kaminska
- 3 Department of Neurology, Medical University of Warsaw , Warsaw, Poland
| | | | - Alicja Jozkowicz
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Agnieszka Loboda
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Jozef Dulak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| |
Collapse
|