1
|
Eyuboglu AA, Akdemir O, Erbas O, Isken MT, Zhang F, Lineaweaver WC. Propionyl-l-carnitine mitigates ischemia-reperfusion injury in rat epigastric island flaps. Heliyon 2024; 10:e27448. [PMID: 38463759 PMCID: PMC10923838 DOI: 10.1016/j.heliyon.2024.e27448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion injury presents a substantial concern in various medical scenarios, notably in reconstructive surgery involving tissue flaps. Despite reports on the protective benefits of Propionyl-l-carnitine against ischemia-reperfusion injury, a thorough assessment of its efficacy in epigastric island flap models is currently lacking. METHODS Sixteen male Sprague-Dawley rats underwent epigastric island flap surgery and were divided into two groups: a Propionyl-l-carnitine group that received intraperitoneal Propionyl-l-carnitine prior to ischemia induction and a sham group that received saline treatment. A comprehensive evaluation was performed including macroscopic, biochemical and histological assessments encompassing measurements of flap survival areas, lipid peroxidation (malondialdehyde), glutathione, myeloperoxidase, nitric oxide and peripheral neutrophil counts. RESULTS The Propionyl-l-carnitine group demonstrated significantly increased flap survival areas when compared to the sham group. Administration of Propionyl-l-carnitine led to reduced malondialdehyde levels and elevated glutathione levels indicating a reduction in oxidative stress. Furthermore, the Propionyl-l-carnitine group exhibited lower myeloperoxidase levels, higher nitric oxide levels and reduced peripheral neutrophil counts, suggesting a decrease in the inflammatory response. Histopathological analysis revealed decreased levels of inflammation, necrosis, polymorphonuclear leukocyte infiltration and edema in the Propionyl-l-carnitine group. Additionally, vascularity was enhanced in the Propionyl-l-carnitine group. CONCLUSION This study provides compelling evidence that Propionyl-l-carnitine administration effectively mitigates the deleterious effects of ischemia-reperfusion injury in epigastric island flaps. This is substantiated by the improved flap survival, diminished oxidative stress and inflammation, as well as the enhanced vascularity observed. Propionyl-l-carnitine emerges as a promising therapeutic intervention to enhance tissue flap survival in reconstructive surgery, warranting further exploration through larger-scale investigations.
Collapse
Affiliation(s)
- Atilla Adnan Eyuboglu
- Arel University, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Ovunc Akdemir
- Aydin University, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Oytun Erbas
- Bilim University, Department of Physiopathology, Istanbul, Turkey
| | - Mustafa Tonguc Isken
- Bahcesehir Medical University, Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Feng Zhang
- PhD University of Mississippi Medical Center, Division of Plastic Surgery, Microsurgery, 2500 North State Street, Jackson, MS 39216, USA
| | - William C. Lineaweaver
- Vanderbilt Bill Wilkerson Center for Otolaryngology and Communication Sciences, Plastic, Reconstructive and Aesthetic Surgery, Nashville, TN, USA
| |
Collapse
|
2
|
Rahmati M, Haffner M, Lee MA, Leach JK, Saiz AM. The critical impact of traumatic muscle loss on fracture healing: Basic science and clinical aspects. J Orthop Res 2024; 42:249-258. [PMID: 37990953 DOI: 10.1002/jor.25746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Musculoskeletal trauma, specifically fractures, is a leading cause of patient morbidity and disability worldwide. In approximately 20% of cases with fracture and related traumatic muscle loss, bone healing is impaired leading to fracture nonunion. Over the past few years, several studies have demonstrated that bone and the surrounding muscle tissue interact not only anatomically and mechanically but also through biochemical pathways and mediators. Severe damage to the surrounding musculature at the fracture site causes an insufficiency in muscle-derived osteoprogenitor cells that are crucial for fracture healing. As an endocrine tissue, skeletal muscle produces many myokines that act on different bone cells, such as osteoblasts, osteoclasts, osteocytes, and mesenchymal stem cells. Investigating how muscle influences fracture healing at cellular, molecular, and hormonal levels provides translational therapeutic solutions to this clinical challenge. This review provides an overview about the contributions of surrounding muscle tissue in directing fracture healing. The focus of the review is on describing the interactions between bone and muscle in both healthy and fractured environments. We discuss current progress in identifying the bone-muscle molecular pathways and strategies to harness these pathways as cues for accelerating fracture healing. In addition, we review the existing challenges and research opportunities in the field.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Max Haffner
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Mark A Lee
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Jonathan Kent Leach
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Augustine M Saiz
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
3
|
Mohamad Yusoff F, Nakashima A, Kajikawa M, Kishimoto S, Maruhashi T, Higashi Y. Therapeutic Myogenesis Induced by Ultrasound Exposure in a Volumetric Skeletal Muscle Loss Injury Model. Am J Sports Med 2023; 51:3554-3566. [PMID: 37743748 DOI: 10.1177/03635465231195850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Low-intensity pulsed ultrasound (LIPUS) irradiation has been shown to induce various responses in different cells. It has been shown that LIPUS activates extracellular signal-regulated kinase 1/2 (ERK1/2) through integrin. PURPOSE To study the effects of LIPUS on myogenic regulatory factors and other related myogenesis elements in a volumetric skeletal muscle loss injury model. STUDY DESIGN Controlled laboratory study. METHODS C57BL/6J mice were subjected to full-thickness muscle defect injury of the quadriceps and treated with direct application of LIPUS 20 min/d or non-LIPUS treatment (control) for 3, 7, and 14 days. LIPUS was also applied to C2C12 cells in culture in the presence of low and high doses of lipopolysaccharides. The expression levels of myogenic regulatory factors and the expression levels of myokine-related and angiogenic-related proteins of the control and LIPUS groups were analyzed. RESULTS Muscle volume in the injury site was restored at day 14 with LIPUS treatment. Paired-box protein 7, myogenic factor 5, myogenin, and desmin expressions were significantly different between control and LIPUS groups at days 7 and 14. Myokine and angiogenic cytokine-related factors were significantly increased in the LIPUS group at day 3 and decreased with no significant difference between the groups by day 14. LIPUS induced different responses of myogenic regulatory factors in C2C12 cells with low and high doses of lipopolysaccharides. LIPUS promoted myogenesis through short-lived increase in interleukin-6 and heme oxygenase 1, together with activation of ERK1/2. CONCLUSION LIPUS had a constant effect on the variables of tissue damage, from macrotrauma to microtrauma, leading to efficient muscle regeneration. CLINICAL RELEVANCE The focus of therapeutic strategies with LIPUS has been not only for microvascular regeneration but also for skeletal muscle and related local tissue recovery from acute or chronic damage.
Collapse
Affiliation(s)
- Farina Mohamad Yusoff
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
4
|
Sun N, Xing Y, Jiang J, Wu P, Qing L, Tang J. Knowledge mapping and emerging trends of ferroptosis in ischemia reperfusion injury research: A bibliometric analysis (2013-2022). Heliyon 2023; 9:e20363. [PMID: 37767486 PMCID: PMC10520329 DOI: 10.1016/j.heliyon.2023.e20363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/31/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE Ischemia/reperfusion (I/R) injury is an inevitable dilemma when previously ischemic multiple organs and tissues are returned to a state of blood flow, with confirming a critical role of ferroptosis in molecular, pathway mechanisms, subcellular structure. Discovering the potential relationship may provide useful approaches for the clinical treatment and prognosis of the pathophysiological status of IRI. Therefore, a comprehensive visualization and scientometric analysis were conducted to systematically summarize and discuss the "ferroptosis in ischemia reperfusion injury" research to demonstrate directions for scholars in this field. METHODS We retrieved all publications focusing on I/R injury and ferroptosis from the Web of Science Core Collection (WoSCC), published from 2013 to October 2022. Next, scientometric analysis of different items was performed using various bibliometrics softwares to explore the annual trends, countries/regions, institutions, journals, authors and their multi-dimensional relationship pointing to current hotspots and future advancement in this field. RESULTS We included a total of 421 English articles in set timespan. The number of publications increased steadily annually. China produced the highest number of publications, followed by the United States. Most publications were from Central South University, followed by Sichuan University and Wuhan University. The most authoritative academic journal was Oxidative Medicine and Cellular Longevity. Cell occupied the first rank of co-cited journal list. Andreas Linkermann and Scott J Dixon may have the highest influence in this intersected field with the highest number of citations and co-cited references respectively. The essential biological reactions such as oxidative stress response, lipid peroxidation metabolism, anti-inflammmatory and pro-inflammatory procedure, and related molecular pathways were knowledge base and current hotspots. Molecules pathways exploration, effective inhibition of I/R injury and promising strategy of improving allografts may become future trends and focuses. CONCLUSIONS Research on ferroptosis in I/R injury had aroused great interest recently. This first bibliometric study comprehensively analyzed the research landscape of ferroptosis and I/R injury, and also provided a reliable reference for related scholars to facilitate further advancement in this field.
Collapse
Affiliation(s)
- Nianzhe Sun
- Department of Orthopedics, Hand & Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yixuan Xing
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Junjie Jiang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Panfeng Wu
- Department of Orthopedics, Hand & Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Liming Qing
- Department of Orthopedics, Hand & Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Juyu Tang
- Department of Orthopedics, Hand & Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
5
|
Tong Z, Li H, Jin Y, Sheng L, Ying M, Liu Q, Wang C, Teng C. Mechanisms of ferroptosis with immune infiltration and inflammatory response in rotator cuff injury. Genomics 2023; 115:110645. [PMID: 37230182 DOI: 10.1016/j.ygeno.2023.110645] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/03/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023]
Abstract
The processes driving ferroptosis and rotator cuff (RC) inflammation are yet unknown. The mechanism of ferroptosis and inflammation involved in the development of RC tears was investigated. The Gene Expression Omnibus database was used to obtain the microarray data relevant to the RC tears for further investigation. In this study, we created an RC tears rat model for in vivo experimental validation. For the additional function enrichment analysis, 10 hub ferroptosis-related genes were chosen to construct the correlation regulation network. In RC tears, it was discovered that genes related to hub ferroptosis and hub inflammatory response were strongly correlated. The outcomes of in vivo tests showed that RC tears were related to Cd68-Cxcl13, Acsl4-Sat1, Acsl3-Eno3, Acsl3-Ccr7, and Ccr7-Eno3 pairings in regulating ferroptosis and inflammatory response. Thus, our results show an association between ferroptosis and inflammation, providing a new avenue to explore the clinical treatment of RC tears.
Collapse
Affiliation(s)
- Zhicheng Tong
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Huimin Li
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Yanglei Jin
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Lingchao Sheng
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Mingshuai Ying
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Qixue Liu
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Chenhuan Wang
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China
| | - Chong Teng
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 32200, China..
| |
Collapse
|
6
|
Zhang D, Tao J, Zhang X, Ma X, Li C, Li H, Li W, Chen J, Liu H. Novel Pro-myogenic Factor Neoruscogenin Induces Muscle Fiber Hypertrophy by Inhibiting MSTN Maturation and Activating the Akt/mTOR Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:499-511. [PMID: 36563293 DOI: 10.1021/acs.jafc.2c06527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neoruscogenin is a plant-origin sapogenin that has the potential to modulate muscle growth among the small-molecule compounds that we previously predicted by artificial intelligence to target myostatin (MSTN). This study aimed to elucidate the biological role of neoruscogenin on muscle growth and its relationship with MSTN. Using molecular biological techniques, we found that neoruscogenin inhibited MSTN maturation, thereby repressing its signal transduction; further facilitated protein synthesis metabolism and reduced protein degradation metabolism, ultimately promoting the differentiation of myoblasts and hypertrophy of muscle fibers; and had the effect of repairing muscle injury. This study enriched the biological functions of neoruscogenin and provided a theoretical basis for the treatment of human myopathy and its application in the livestock industry.
Collapse
Affiliation(s)
- Dingding Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingli Tao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangfei Ma
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chengyu Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hongmin Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weijian Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
7
|
Kelestemur T, Nemeth Z, Pacher P, Antonioli L, Haskó G. A 2A ADENOSINE RECEPTORS REGULATE MULTIPLE ORGAN FAILURE AFTER HEMORRHAGIC SHOCK IN MICE. Shock 2022; 58:321-331. [PMID: 36018304 PMCID: PMC10292675 DOI: 10.1097/shk.0000000000001985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Trauma hemorrhagic shock (T/HS) is a clinical condition that causes multiple organ failure that needs rapid intervention. Restricted oxygen at the cellular level causes inflammation and subsequent cell death. Adenosine triphosphate is the universal intracellular energy currency and an important extracellular inflammatory signaling molecule. Adenosine, an endogenous nucleotide formed as a result of the breakdown of adenosine triphosphate, is also released during T/HS. Adenosine binds to four G protein-coupled receptors (A 1R , A 2a , A 2b , A 3R ) called adenosine receptors or P1 receptors. In the present study, we evaluated the effect of activation, inactivation, and genetic absence of A2aR (A2aR -/- mice) on T/HS-induced multiple organ failure. Wild-type mice were pretreated (30 min before shock induction) with an agonist or antagonist and then subjected to T/HS by withdrawing arterial blood and maintaining the blood pressure between 28 and 32 mm Hg. A2aR -/- mice were subjected to T/HS in the absence of pharmacologic treatment. Neutrophil sequestration was assessed by detecting myeloperoxidase, and Evans blue dye (EBD) method was used to analyze lung permeability. Blood and lung inflammatory cytokine levels were determined by sandwich enzyme-linked immunosorbent assay. The liver enzymes aspartate aminotransferase and alanine aminotransferase were determined spectrophotometrically from plasma. Activation of the apoptotic cascade was evaluated using a mouse apoptosis array. Our results demonstrate that the selective A2aR agonist CGS21680 decreases lung neutrophil sequestration, lung proinflammatory cytokines IL-6 and TNF-α, and bronchoalveolar lavage EBD. Pretreatment with the selective antagonist ZM241385 and genetic blockade in A2aR -/- mice increased neutrophil sequestration, proinflammatory cytokine levels, and bronchoalveolar lavage fluid EBD. The myeloperoxidase level in the lung was also increased in A2aR -/- mice. We observed that antiapoptotic markers decreased significantly with the absence of A2aR in the lung and spleen after T/HS. In conclusion, our data demonstrate that activation of A2aR regulates organ injury and apoptosis in the setting of T/HS.
Collapse
Affiliation(s)
- Taha Kelestemur
- Department of Anesthesiology, Columbia University, NY 10032, USA
- Department of Physiology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkiye
| | - Zoltan Nemeth
- Department of Anesthesiology, Columbia University, NY 10032, USA
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
| | - Pal Pacher
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - György Haskó
- Department of Anesthesiology, Columbia University, NY 10032, USA
| |
Collapse
|
8
|
Drysch M, Schmidt SV, Becerikli M, Reinkemeier F, Dittfeld S, Wagner JM, Dadras M, Sogorski A, von Glinski M, Lehnhardt M, Behr B, Wallner C. Myostatin Deficiency Protects C2C12 Cells from Oxidative Stress by Inhibiting Intrinsic Activation of Apoptosis. Cells 2021; 10:cells10071680. [PMID: 34359850 PMCID: PMC8305813 DOI: 10.3390/cells10071680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/09/2023] Open
Abstract
Ischemia reperfusion (IR) injury remains an important topic in clinical medicine. While a multitude of prophylactic and therapeutic strategies have been proposed, recent studies have illuminated protective effects of myostatin inhibition. This study aims to elaborate on the intracellular pathways involved in myostatin signaling and to explore key proteins that convey protective effects in IR injury. We used CRISPR/Cas9 gene editing to introduce a myostatin (Mstn) deletion into a C2C12 cell line. In subsequent experiments, we evaluated overall cell death, activation of apoptotic pathways, ROS generation, lipid peroxidation, intracellular signaling via mitogen-activated protein kinases (MAPKs), cell migration, and cell proliferation under hypoxic conditions followed by reoxygenation to simulate an IR situation in vitro (hypoxia reoxygenation). It was found that mitogen-activated protein kinase kinase 3/6, also known as MAPK/ERK Kinase 3/6 (MEK3/6), and subsequent p38 MAPK activation were blunted in C2C12-Mstn−/− cells in response to hypoxia reoxygenation (HR). Similarly, c-Jun N-terminal kinase (JNK) activation was negated. We also found the intrinsic activation of apoptosis to be more important in comparison with the extrinsic activation. Additionally, intercepting myostatin signaling mitigated apoptosis activation. Ultimately, this research validated protective effects of myostatin inhibition in HR and identified potential mediators worth further investigation. Intercepting myostatin signaling did not inhibit ROS generation overall but mitigated cellular injury. In particular, intrinsic activation of apoptosis origination from mitochondria was alleviated. This was presumably mediated by decreased activation of p38 caused by the diminished kinase activity increase of MEK3/6. Overall, this work provides important insights into HR signaling in C2C12-Mstn−/− cells and could serve as basis for further research.
Collapse
|