1
|
Yang L, Huang H, Fan Y, Xu L, Jin X, Xiao B, Ma C, Fan H, Chai Z. Astragaloside IV ameliorates Parkinson's disease by inhibiting TLR4/NF-κB-dependent neuroinflammation. Int Immunopharmacol 2025; 160:114972. [PMID: 40449268 DOI: 10.1016/j.intimp.2025.114972] [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: 01/19/2025] [Revised: 05/26/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025]
Abstract
Astragaloside IV (AS-IV) is a bioactive compound derived from Radix Astragali, a traditional Chinese herb widely used as a dietary supplement to enhance immune function. Modern pharmacological studies have demonstrated that AS-IV exhibits anti-inflammatory and immunomodulatory properties. In this study, we investigated the effects of AS-IV on motor dysfunction, microglial polarization, and immune regulation mechanisms in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) mouse model. Our results showed that AS-IV (40 mg/kg) significantly improved motor function in PD mice, as evidenced by reduced descent time in the pole test, increased hanging score in the hanging test, increased stride lengths, and reduced paw angle in the gait test. Furthermore, AS-IV administration attenuated the loss of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra pars compacta (SNpc), promoted microglial polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, suppressed the levels of pro-inflammatory cytokines including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNFα), enhanced the levels of anti-inflammatory cytokines including interleukin-4 (IL-4) and interleukin-10 (IL-10) in the SNpc of PD mice. Mechanistically, AS-IV significantly downregulated the expression and phosphorylation levels of TLR4/p38 (JNK)/NF-κB pathway-related proteins, including Toll-like receptor 4 (TLR4), Myeloid differentiation primary response protein 88 (MyD88), Apoptosis signal-regulating kinase 1 (ASK1), Mitogen-activated protein kinase 3/6 (MKK3/6), Phosphorylated-MKK3/6 (p-MKK3/6), Phosphorylated-mitogen-activated protein kinase 4/7 (p-MKK4/7), p38 mitogen-activated protein kinase (p38), Phosphorylated-p38 (p-p38), c-Jun N-terminal kinase (JNK), Phosphorylated-JNK (p-JNK), nuclear factor kappa-B (NF-κB), and Phosphorylated-NF-κB (p-NF-κB). To further validate the targeting effect of AS-IV, 1 mg/kg of LPS-EB Ultrapure was utilized as a specific TLR4 agonistwe to selectively activated the TLR4/NF-κB signaling pathway without triggering other inflammatory pathways, leading to elevated mRNA levels of TLR4, NF-κB, IL-1β, IL-6, TNFα and protein expression of TLR4, p-JNK, p-p38, p-NF-κB, IL-1β, IL-6, TNFα in the SNpc of PD mice. Importantly, AS-IV pretreatment can't counteract these LPS-EB Ultrapure-triggered effects, demonstrating its dependence on the TLR4/NF-κB signaling pathway. In conclusion, our findings indicate that AS-IV modulates microglial polarization and attenuates neuroinflammation by inhibiting the TLR4/NF-κB pathway, thereby ameliorating motor dysfunction and neuronal loss in PD mice.
Collapse
Affiliation(s)
- Lixia Yang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, China; The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Haihua Huang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, China; The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Yue Fan
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Lei Xu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Xiaoming Jin
- Department of Anatomy and Cell Biology, Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Baoguo Xiao
- Huashan Hospital, Fudan University, Shanghai 200025, China
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Huijie Fan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Zhi Chai
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
2
|
Lin L, Wei J, Xue J, Fan G, Zhu W, Zhu Y, Wu R. Drp1 Promotes Macrophage M1 Polarization and Inflammatory Response in Autoimmune Myocarditis by Driving Mitochondrial Fission. J Cardiovasc Transl Res 2025; 18:237-246. [PMID: 39388091 DOI: 10.1007/s12265-024-10570-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Autoimmune myocarditis (AM) is characterized by an intricate inflammatory response within the myocardium. Dynamin-related protein 1 (Drp1), a pivotal modulator of mitochondrial fission, plays a role in the pathogenesis of various diseases. A myosin-induced experimental autoimmune myocarditis (EAM) mouse model was successfully established. Flow cytometry was employed to detect M1/M2-like macrophages. Mitochondrial fragmentation was assessed using Mito-Tracker Red CMXRos. Drp1 was upregulated and activated in EAM mice. Depletion of Drp1 was observed to mitigate inflammation, macrophage infiltration and M1 polarization within the cardiac tissue of EAM mice. In M1-like macrophages derived from the hearts of EAM mice, Drp1 was found to promote mitochondrial fission and diminish mitochondrial fusion. Furthermore, the depletion of Drp1 reduced the NF-κB-related pro-inflammatory response in EAM-associated M1-like macrophages. Drp1 drives mitochondrial fission in macrophages, driving their M1 polarization and the subsequent inflammatory response. Drp1 may represent an effective target for the prevention and treatment of AM.
Collapse
Affiliation(s)
- Lin Lin
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China.
| | - Jin Wei
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Jiahong Xue
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Gang Fan
- Second Department of Cardiology, Xianyang First People's Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Wenjing Zhu
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Yanhe Zhu
- Institute of Endiquidiopathies, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruiyun Wu
- Department of Internal Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| |
Collapse
|
3
|
Vicenzetto C, Giordani AS, Menghi C, Baritussio A, Scognamiglio F, Pontara E, Bison E, Peloso-Cattini MG, Marcolongo R, Caforio ALP. Cellular Immunology of Myocarditis: Lights and Shades-A Literature Review. Cells 2024; 13:2082. [PMID: 39768171 PMCID: PMC11674465 DOI: 10.3390/cells13242082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium with heterogeneous etiology, clinical presentation, and prognosis; when it is associated with myocardial dysfunction, this identifies the entity of inflammatory cardiomyopathy. In the last few decades, the relevance of the immune system in myocarditis onset and progression has become evident, thus having crucial clinical relevance in terms of treatment and prognostic stratification. In fact, the advances in cardiac immunology have led to a better characterization of the cellular subtypes involved in the pathogenesis of inflammatory cardiomyopathy, whether the etiology is infectious or autoimmune/immune-mediated. The difference in the clinical course between spontaneous recovery to acute, subacute, or chronic progression to end-stage heart failure may be explained not only by classical prognostic markers but also through immune-pathological mechanisms at a cellular level. Nevertheless, much still needs to be clarified in terms of immune characterization and molecular mechanisms especially in biopsy-proven myocarditis. The aims of this review are to (1) describe inflammatory cardiomyopathy etiology, especially immune-mediated/autoimmune forms, (2) analyze recent findings on the role of different immune cells subtypes in myocarditis, (3) illustrate the potential clinical relevance of such findings, and (4) highlight the need of further studies in pivotal areas of myocarditis cellular immunology.
Collapse
Affiliation(s)
- Cristina Vicenzetto
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Caterina Menghi
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anna Baritussio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Federico Scognamiglio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elena Pontara
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elisa Bison
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Maria Grazia Peloso-Cattini
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Renzo Marcolongo
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
4
|
Fang Y, Min S, Shen H. The role of mitochondrial fusion and fission in immune-mediated inflammatory diseases. Cell Immunol 2024; 403-404:104864. [PMID: 39190985 DOI: 10.1016/j.cellimm.2024.104864] [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: 06/17/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
Mitochondria are highly dynamic organelles that maintain their homeostasis through mitochondrial dynamics. Mitochondrial fusion and fission are two important processes of mitochondrial dynamics. There is accumulating evidence that mitochondrial fusion and fission play an important role in the development of immune-mediated inflammatory diseases. This article provides a brief review of the essential role of mitochondrial fusion and fission in immune-mediated inflammatory diseases. It will provide a novel perspective and direction for the elucidation of the pathogenesis and treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yulai Fang
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shichen Min
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hong Shen
- Nanjing University of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
5
|
Huang Q, Chen T, Li J, Wang Y, Shi H, Yu Y, Ji Q, Shen X, Sun T, Shi H, Luo X, Jin B, You Y, Wu B. IL-37 ameliorates myocardial fibrosis by regulating mtDNA-enriched vesicle release in diabetic cardiomyopathy mice. J Transl Med 2024; 22:494. [PMID: 38790051 PMCID: PMC11127460 DOI: 10.1186/s12967-024-05250-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM), a serious complication of diabetes, leads to structural and functional abnormalities of the heart and ultimately evolves to heart failure. IL-37 exerts a substantial influence on the regulation of inflammation and metabolism. Whether IL-37 is involved in DCM is unknown. METHODS The plasma samples were collected from healthy controls, diabetic patients and DCM patients, and the level of IL-37 and its relationship with heart function were observed. The changes in cardiac function, myocardial fibrosis and mitochondrial injury in DCM mice with or without IL-37 intervention were investigated in vivo. By an in vitro co-culture approach involving HG challenge of cardiomyocytes and fibroblasts, the interaction carried out by cardiomyocytes on fibroblast profibrotic activation was studied. Finally, the possible interactive mediator between cardiomyocytes and fibroblasts was explored, and the intervention role of IL-37 and its relevant molecular mechanisms. RESULTS We showed that the level of plasma IL-37 in DCM patients was upregulated compared to that in healthy controls and diabetic patients. Both recombinant IL-37 administration or inducing IL-37 expression alleviated cardiac dysfunction and myocardial fibrosis in DCM mice. Mechanically, hyperglycemia impaired mitochondria through SIRT1/AMPK/PGC1α signaling, resulting in significant cardiomyocyte apoptosis and the release of extracellular vesicles containing mtDNA. Fibroblasts then engulfed these mtDNA-enriched vesicles, thereby activating TLR9 signaling and the cGAS-STING pathway to initiate pro-fibrotic process and adverse remodeling. However, the presence of IL-37 ameliorated mitochondrial injury by preserving the activity of SIRT1-AMPK-PGC1α axis, resulting in a reduction in release of mtDNA-enriched vesicle and ultimately attenuating the progression of DCM. CONCLUSIONS Collectively, our study demonstrates a protective role of IL-37 in DCM, offering a promising therapeutic agent for this disease.
Collapse
Affiliation(s)
- Qingyu Huang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongqing Chen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Jian Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiming Wang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huairui Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifei Yu
- Endocrinology department, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoyan Shen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Tao Sun
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haiming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinping Luo
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bo Jin
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yan You
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China.
| | - Bangwei Wu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Ray AK, Priya A, Malik MZ, Thanaraj TA, Singh AK, Mago P, Ghosh C, Shalimar, Tandon R, Chaturvedi R. A bioinformatics approach to elucidate conserved genes and pathways in C. elegans as an animal model for cardiovascular research. Sci Rep 2024; 14:7471. [PMID: 38553458 PMCID: PMC10980734 DOI: 10.1038/s41598-024-56562-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/07/2024] [Indexed: 04/02/2024] Open
Abstract
Cardiovascular disease (CVD) is a collective term for disorders of the heart and blood vessels. The molecular events and biochemical pathways associated with CVD are difficult to study in clinical settings on patients and in vitro conditions. Animal models play a pivotal and indispensable role in CVD research. Caenorhabditis elegans, a nematode species, has emerged as a prominent experimental organism widely utilized in various biomedical research fields. However, the specific number of CVD-related genes and pathways within the C. elegans genome remains undisclosed to date, limiting its in-depth utilization for investigations. In the present study, we conducted a comprehensive analysis of genes and pathways related to CVD within the genomes of humans and C. elegans through a systematic bioinformatic approach. A total of 1113 genes in C. elegans orthologous to the most significant CVD-related genes in humans were identified, and the GO terms and pathways were compared to study the pathways that are conserved between the two species. In order to infer the functions of CVD-related orthologous genes in C. elegans, a PPI network was constructed. Orthologous gene PPI network analysis results reveal the hubs and important KRs: pmk-1, daf-21, gpb-1, crh-1, enpl-1, eef-1G, acdh-8, hif-1, pmk-2, and aha-1 in C. elegans. Modules were identified for determining the role of the orthologous genes at various levels in the created network. We also identified 9 commonly enriched pathways between humans and C. elegans linked with CVDs that include autophagy (animal), the ErbB signaling pathway, the FoxO signaling pathway, the MAPK signaling pathway, ABC transporters, the biosynthesis of unsaturated fatty acids, fatty acid metabolism, glutathione metabolism, and metabolic pathways. This study provides the first systematic genomic approach to explore the CVD-associated genes and pathways that are present in C. elegans, supporting the use of C. elegans as a prominent animal model organism for cardiovascular diseases.
Collapse
Affiliation(s)
- Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| | - Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait.
| | | | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Science for Women, University of Delhi, New Delhi, India
- Campus of Open Learning, University of Delhi, New Delhi, India
| | - Chirashree Ghosh
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
7
|
Ray AK, Priya A, Malik MZ, Thanaraj TA, Singh AK, Mago P, Ghosh C, Shalimar, Tandon R, Chaturvedi R. Conserved Cardiovascular Network: Bioinformatics Insights into Genes and Pathways for Establishing Caenorhabditis elegans as an Animal Model for Cardiovascular Diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.24.573256. [PMID: 38234826 PMCID: PMC10793405 DOI: 10.1101/2023.12.24.573256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Cardiovascular disease (CVD) is a collective term for disorders of the heart and blood vessels. The molecular events and biochemical pathways associated with CVD are difficult to study in clinical settings on patients and in vitro conditions. Animal models play a pivotal and indispensable role in cardiovascular disease (CVD) research. Caenorhabditis elegans , a nematode species, has emerged as a prominent experimental organism widely utilised in various biomedical research fields. However, the specific number of CVD-related genes and pathways within the C. elegans genome remains undisclosed to date, limiting its in-depth utilisation for investigations. In the present study, we conducted a comprehensive analysis of genes and pathways related to CVD within the genomes of humans and C. elegans through a systematic bioinformatic approach. A total of 1113 genes in C. elegans orthologous to the most significant CVD-related genes in humans were identified, and the GO terms and pathways were compared to study the pathways that are conserved between the two species. In order to infer the functions of CVD-related orthologous genes in C. elegans, a PPI network was constructed. Orthologous gene PPI network analysis results reveal the hubs and important KRs: pmk-1, daf-21, gpb-1, crh-1, enpl-1, eef-1G, acdh-8, hif-1, pmk-2, and aha-1 in C. elegans. Modules were identified for determining the role of the orthologous genes at various levels in the created network. We also identified 9 commonly enriched pathways between humans and C. elegans linked with CVDs that include autophagy (animal), the ErbB signalling pathway, the FoxO signalling pathway, the MAPK signalling pathway, ABC transporters, the biosynthesis of unsaturated fatty acids, fatty acid metabolism, glutathione metabolism, and metabolic pathways. This study provides the first systematic genomic approach to explore the CVD-associated genes and pathways that are present in C. elegans, supporting the use of C. elegans as a prominent animal model organism for cardiovascular diseases.
Collapse
|
8
|
Liu C, Xu X, He X, Ren J, Chi M, Deng G, Li G, Nasser MI. Activation of the Nrf-2/HO-1 signalling axis can alleviate metabolic syndrome in cardiovascular disease. Ann Med 2023; 55:2284890. [PMID: 38039549 PMCID: PMC10836253 DOI: 10.1080/07853890.2023.2284890] [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: 05/28/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
Background: Cardiovascular disease (CVD) is widely observed in modern society. CVDs are responsible for the majority of fatalities, with heart attacks and strokes accounting for approximately 80% of these cases. Furthermore, a significant proportion of these deaths, precisely one-third, occurs in individuals under 70. Metabolic syndrome encompasses a range of diseases characterized by various physiological dysfunctions. These include increased inflammation in adipose tissue, enhanced cholesterol synthesis in the liver, impaired insulin secretion, insulin resistance, compromised vascular tone and integrity, endothelial dysfunction, and atheroma formation. These factors contribute to the development of metabolic disorders and significantly increase the likelihood of experiencing cardiovascular complications.Method: We selected studies that proposed hypotheses regarding metabolic disease syndrome and cardiovascular disease (CVD) and the role of Nrf2/HO-1 and factor regulation in CVD research investigations based on our searches of Medline and PubMed.Results: A total of 118 articles were included in the review, 16 of which exclusively addressed hypotheses about the role of Nrf2 on Glucose regulation, while 16 involved Cholesterol regulation. Likewise, 14 references were used to prove the importance of mitochondria on Nrf2. Multiple studies have provided evidence suggesting the involvement of Nrf2/HO-1 in various physiological processes, including metabolism and immune response. A total of 48 research articles and reviews have been used to highlight the role of metabolic syndrome and CVD.Conclusion: This review provides an overview of the literature on Nrf2/HO-1 and its role in metabolic disease syndrome and CVD.
Collapse
Affiliation(s)
- Chi Liu
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xingli Xu
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingxuan Chi
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Gang Deng
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Cardiovascular Institute, Guangzhou, Guangdong, China
| | - Guisen Li
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Moussa Ide Nasser
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Cardiovascular Institute, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Chen J, Shao J, Wang Y, Wu K, Huang M. OPA1, a molecular regulator of dilated cardiomyopathy. J Cell Mol Med 2023; 27:3017-3025. [PMID: 37603376 PMCID: PMC10568666 DOI: 10.1111/jcmm.17918] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/22/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a disease with no specific treatment, poor prognosis and high mortality. During DCM development, there is apoptosis, mitochondrial dynamics imbalance and changes in cristae structure. Optic atrophy 1 (OPA1) appears at high frequency in these three aspects. DCM LMNA (LaminA/C) gene mutation can activate TP53, and the study of P53 shows that P53 affects OPA1 through Bak/Bax and OMA1 (a metalloprotease). OPA1 can be considered the missing link between DCMp53 and DCM apoptosis, mitochondrial dynamics imbalance and changes in cristae structure. OPA1 regulates apoptosis by regulating the release of cytochrome c from the mitochondrial matrix through CJs (crisp linkages, located in the inner mitochondrial membrane) and unbalances mitochondrial fusion and fission by affecting mitochondrial inner membrane (IM) fusion. OPA1 is also associated with the formation and maintenance of mitochondrial cristae. OPA1 is not the root cause of DCM, but it is an essential mediator in P53 mediating the occurrence and development of DCM, so OPA1 also becomes a molecular regulator of DCM. This review discusses the implication of OPA1 for DCM from three aspects: apoptosis, mitochondrial dynamics and ridge structure.
Collapse
Affiliation(s)
- Jiaqi Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jianan Shao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yaoyao Wang
- Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
| | - Kangxiang Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Mingyuan Huang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
10
|
Hu J, Xu J, Tan X, Li D, Yao D, Xu B, Lei Y. Dapagliflozin protects against dilated cardiomyopathy progression by targeting NLRP3 inflammasome activation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1461-1470. [PMID: 36749400 PMCID: PMC10244283 DOI: 10.1007/s00210-023-02409-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/26/2023] [Indexed: 02/08/2023]
Abstract
Dilated cardiomyopathy (DCM) is the major cause of heart failure and has a poor prognosis. The accumulating evidence points to an essential role of the inflammatory component in the process of DCM. Inhibitors of sodium-glucose cotransporter 2 (SGLT2) are widely used to treat heart failure patients due to their cardiac benefits. However, their role in DCM remains unclear. We used the doxorubicin (Dox)-induced DCM model for our study. The SGLT2 inhibitor dapagliflozin (Dapa) improved cardiac function in mice treated with doxorubicin and attenuated the activation of the nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome pathway and the expression of inflammatory factors. In addition, dapagliflozin suppresses NLRP3 activation by decreasing p38-dependent toll-like receptor 4 (TLR4) expression. In our study, dagliflozin improves cardiac function in DCM by inhibiting the activity of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No.158 Wuyang Avenue, Enshi, 445000, Hubei, China
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiamin Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xi Tan
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Dong Li
- Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Enshi, Hubei, China
| | - Dejiang Yao
- Surgical Division IIIThe Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China.
| | - Yuhua Lei
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No.158 Wuyang Avenue, Enshi, 445000, Hubei, China.
| |
Collapse
|
11
|
Castro-Sepulveda M, Tuñón-Suárez M, Rosales-Soto G, Vargas-Foitzick R, Deldicque L, Zbinden-Foncea H. Regulation of mitochondrial morphology and cristae architecture by the TLR4 pathway in human skeletal muscle. Front Cell Dev Biol 2023; 11:1212779. [PMID: 37435031 PMCID: PMC10332154 DOI: 10.3389/fcell.2023.1212779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
In skeletal muscle (SkM), a reduced mitochondrial elongate phenotype is associated with several metabolic disorders like type 2 diabetes mellitus (T2DM). However, the mechanisms contributing to this reduction in mitochondrial elongate phenotype in SkM have not been fully elucidated. It has recently been shown in a SkM cell line that toll-like receptor 4 (TLR4) contributes to the regulation of mitochondrial morphology. However, this has not been investigated in human SkM. Here we found that in human SkM biopsies, TLR4 protein correlated negatively with Opa1 (pro-mitochondrial fusion protein). Moreover, the incubation of human myotubes with LPS reduced mitochondrial size and elongation and induced abnormal mitochondrial cristae, which was prevented with the co-incubation of LPS with TAK242. Finally, T2DM myotubes were found to have reduced mitochondrial elongation and mitochondrial cristae density. Mitochondrial morphology, membrane structure, and insulin-stimulated glucose uptake were restored to healthy levels in T2DM myotubes treated with TAK242. In conclusion, mitochondrial morphology and mitochondrial cristae seem to be regulated by the TLR4 pathway in human SkM. Those mitochondrial alterations might potentially contribute to insulin resistance in the SkM of patients with T2DM.
Collapse
Affiliation(s)
- Mauricio Castro-Sepulveda
- Laboratorio de Fisiología del Ejercicio y Metabolismo, Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Mauro Tuñón-Suárez
- Laboratorio de Fisiología del Ejercicio y Metabolismo, Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Giovanni Rosales-Soto
- Facultad de Ciencias de la Educación, Universidad San Sebastián, Sede Bellavista, Santiago, Chile
| | - Ronald Vargas-Foitzick
- Laboratorio de Fisiología del Ejercicio y Metabolismo, Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Louise Deldicque
- Institute of Neuroscience, UCLouvain, Ottignies-Louvain-la- Neuve, Belgium
| | - Hermann Zbinden-Foncea
- Laboratorio de Fisiología del Ejercicio y Metabolismo, Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
- Institute of Neuroscience, UCLouvain, Ottignies-Louvain-la- Neuve, Belgium
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Madrid, España
| |
Collapse
|
12
|
Gao J, Hou T. Cardiovascular disease treatment using traditional Chinese medicine:Mitochondria as the Achilles' heel. Biomed Pharmacother 2023; 164:114999. [PMID: 37311280 DOI: 10.1016/j.biopha.2023.114999] [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: 04/08/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
Cardiovascular disease (CVD), involving the pathological alteration of the heart or blood vessels, is one of the main causes of disability and death worldwide, with an estimated 18.6 million deaths per year. CVDs are caused by a variety of risk factors, including inflammation, hyperglycemia, hyperlipidemia, and increased oxidative stress. Mitochondria, the hub of ATP production and the main generator of reactive oxygen species (ROS), are linked to multiple cellular signaling pathways that regulate the progression of CVD and therefore are recognized as an essential target for CVD management. Initial treatment of CVD generally focuses on diet and lifestyle interventions; proper drugs or surgery can prolong or save the patient's life. Traditional Chinese medicine (TCM), a holistic medical care system with an over 2500-year history, has been proven to be efficient in curing CVD and other illnesses, with a strengthening effect on the body. However, the mechanisms underlying TCM alleviation of CVD remain elusive. Recent studies have recognized that TCM can alleviate cardiovascular disease by manipulating the quality and function of mitochondria. This review systematically summarizes the association of mitochondria with cardiovascular risk factors, and the relationships between mitochondrial dysfunction and CVD progression. We will investigate the research progress of managing cardiovascular disease by TCM and cover widely used TCMs that target mitochondria for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Jie Gao
- Chengdu Integrated TCM and Western Medicine Hospital and Chengdu University of Traditional Chinese Medicine, Chengdu 610041 China
| | - Tianshu Hou
- Chengdu Integrated TCM and Western Medicine Hospital and Chengdu University of Traditional Chinese Medicine, Chengdu 610041 China.
| |
Collapse
|
13
|
Gawali B, Sridharan V, Krager KJ, Boerma M, Pawar SA. TLR4-A Pertinent Player in Radiation-Induced Heart Disease? Genes (Basel) 2023; 14:genes14051002. [PMID: 37239362 DOI: 10.3390/genes14051002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The heart is one of the organs that is sensitive to developing delayed adverse effects of ionizing radiation (IR) exposure. Radiation-induced heart disease (RIHD) occurs in cancer patients and cancer survivors, as a side effect of radiation therapy of the chest, with manifestation several years post-radiotherapy. Moreover, the continued threat of nuclear bombs or terrorist attacks puts deployed military service members at risk of exposure to total or partial body irradiation. Individuals who survive acute injury from IR will experience delayed adverse effects that include fibrosis and chronic dysfunction of organ systems such as the heart within months to years after radiation exposure. Toll-like receptor 4 (TLR4) is an innate immune receptor that is implicated in several cardiovascular diseases. Studies in preclinical models have established the role of TLR4 as a driver of inflammation and associated cardiac fibrosis and dysfunction using transgenic models. This review explores the relevance of the TLR4 signaling pathway in radiation-induced inflammation and oxidative stress in acute as well as late effects on the heart tissue and the potential for the development of TLR4 inhibitors as a therapeutic target to treat or alleviate RIHD.
Collapse
Affiliation(s)
- Basveshwar Gawali
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kimberly J Krager
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Snehalata A Pawar
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
14
|
Yang J, Yang K, Wang K, Zhou D, Zhou J, Du X, Liu S, Cheng Z. Serum amyloid A regulates TLR2/4-mediated IFN-β signaling pathway against Marek's disease virus. Virus Res 2023; 326:199044. [PMID: 36652973 DOI: 10.1016/j.virusres.2023.199044] [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: 10/12/2022] [Revised: 01/09/2023] [Accepted: 01/13/2023] [Indexed: 01/16/2023]
Abstract
Serum amyloid A (SAA), an acute response phase protein (APP), is crucial for the innate immune response during pathogenic microorganisms' invasion. Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that activates multiple innate immune molecules, including SAA, in the host during infection. However, the pathway through which SAA participates in MDV-induced host innate immunity remains unknown. The present study aimed to elucidate the pathway through which SAA exerts its anti-MDV function. We observed that MDV infection in vivo and in vitro significantly elevated SAA expression. Furthermore, through SAA overexpression and knockdown experiments, we demonstrated that SAA could inhibit MDV replication. Subsequently, we found that SAA activated Toll-Like Receptor 2/4 (TLR2/4) -mediated Interferon Beta (IFN-β) promoter activity and IFN regulatory factor 7 (IRF7) promoter activity. During MDV infection, SAA enhanced TLR2/4-mediated IFN-β signal transduction and messenger RNAs (mRNAs) expression of type I IFN (IFN-I) and interferon-stimulated genes (ISGs). Finally, TLR2/4 inhibitor OxPAPC inhibits the anti-MDV activity of SAA. These results demonstrated that SAA inhibits MDV replication and enhancing TLR2/4-mediated IFN-β signal transduction to promote IFNs and ISGs expression. This finding is the first to demonstrate the signaling pathway by which SAA exerts its anti-MDV function. It also provides new insights into the control of oncogenic herpesviruses from the perspective of acute response phase proteins.
Collapse
Affiliation(s)
- Jianhao Yang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Kunmei Yang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Kang Wang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Jing Zhou
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Xusheng Du
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Shenglong Liu
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China.
| |
Collapse
|
15
|
Ma J, Luo Y, Liu Y, Chen C, Chen A, Liang L, Wang W, Song Y. Exosome-mediated lnc-ABCA12-3 promotes proliferation and glycolysis but inhibits apoptosis by regulating the toll-like receptor 4/nuclear factor kappa-B signaling pathway in esophageal squamous cell carcinoma. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:61-73. [PMID: 36575934 PMCID: PMC9806635 DOI: 10.4196/kjpp.2023.27.1.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 12/29/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a kind of malignant tumor with high incidence and mortality in the digestive system. The aim of this study is to explore the function of lnc-ABCA12-3 in the development of ESCC and its unique mechanisms. RT-PCR was applied to detect gene transcription levels in tissues or cell lines like TE-1, EC9706, and HEEC cells. Western blot was conducted to identify protein expression levels of mitochondrial apoptosis and toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway. CCK-8 and EdU assays were carried out to measure cell proliferation, and cell apoptosis was examined by flow cytometry. ELISA was used for checking the changes in glycolysis-related indicators. Lnc-ABCA12-3 was highly expressed in ESCC tissues and cells, which preferred it to be a candidate target. The TE-1 and EC9706 cells proliferation and glycolysis were obviously inhibited with the downregulation of lnc-ABCA12-3, while apoptosis was promoted. TLR4 activator could largely reverse the apoptosis acceleration and relieved the proliferation and glycolysis suppression caused by lnc-ABCA12-3 downregulation. Moreover, the effect of lnc-ABCA12-3 on ESCC cells was actualized by activating the TLR4/NF-κB signaling pathway under the mediation of exosome. Taken together, the lnc-ABCA12-3 could promote the proliferation and glycolysis of ESCC, while repressing its apoptosis probably by regulating the TLR4/NF-κB signaling pathway under the mediation of exosome.
Collapse
Affiliation(s)
- Junliang Ma
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China,Correspondence Junliang Ma, E-mail:
| | - Yijun Luo
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yingjie Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Zuinyi Medical University, Zunyi, Guizhou 563003, China
| | - Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Anping Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lubiao Liang
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Wenxiang Wang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410031, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| |
Collapse
|
16
|
Guo H, Xie M, Liu W, Chen S, Ye B, Yao J, Xiao Z, Zhou C, Zheng M. Inhibition of BTK improved APAP-induced liver injury via suppressing proinflammatory macrophages activation by restoring mitochondrion function. Int Immunopharmacol 2022; 110:109036. [PMID: 35850053 DOI: 10.1016/j.intimp.2022.109036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Acetaminophen (APAP) overdose can cause severe liver injury and APAP-induced liver injury (AILI) is one of the leading causes of acute liver failure (ALF). Bruton's tyrosine kinase (BTK) is a key tyrosine kinase in immune responses, which plays an important role in many inflammatory diseases. However, its effect on AILI is still not clear. Here, we aimed to assess the effect of BTK on AILI and explore its underlying mechanism. METHODS In our study, western blot and immunohistochemistry were used to detect the expression of BTK in AILI. The C57BL/6 mice were used to check the protective effect of BTK inhibition on AILI and the activation of BTK was confirmed in mice macrophages treated with APAP. Immunofluorescence, immunohistochemistry, oxygen consumption rate (OCR) detection, polymerase chain reaction (PCR), flow cytometry and western blot were used to determine the role of BTK in mitochondrial dynamics and function of macrophages and the underlying mechanisms in AILI. RESULTS Our results showed that BTK upregulated in AILI. BTK inhibition protected mice from AILI and BTK was activated in mice macrophages in response to APAP. Mechanically, BTK inhibition promoted mitochondrial fusion and restored mitochondrial function through phospholipase C gamma 2 (PLCγ2)-reactive oxygen species (ROS)-Optic Atrophy 1(OPA1) pathway in macrophages and finally suppressed the release of proinflammatory cytokines. CONCLUSIONS In conclusion, we found that BTK inhibition protected mice from AILI by restoring the mitochondrial function of macrophages through the improvement of the mitochondrial dynamic imbalance via PLCγ2-ROS-OPA1 signaling pathway, which indicated that BTK might be a potential therapeutic target of AILI.
Collapse
Affiliation(s)
- Huiting Guo
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Mingjie Xie
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Weixia Liu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Shiwei Chen
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Bingjue Ye
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jiping Yao
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zhengyun Xiao
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Cheng Zhou
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.
| | - Min Zheng
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.
| |
Collapse
|
17
|
TLR4-SIRT3 Mechanism Modulates Mitochondrial and Redox Homeostasis and Promotes EPCs Recruitment and Survival. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1282362. [PMID: 35832490 PMCID: PMC9273456 DOI: 10.1155/2022/1282362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
The low survival rate of endothelial progenitor cells (EPCs) in vivo which are susceptible to adverse microenvironments including inflammation and oxidative stress has become one primary challenge of EPCs transplantation for regenerative therapy. Recent studies reported functional expression of toll-like receptor (TLR) 4 on EPCs and dose-dependent effects of lipopolysaccharide (LPS) on cellular oxidative stress and angiogenic properties. However, the involved mechanism has not yet been elucidated well, and the influence of TLR4 signaling on EPCs survival and function in vivo is unknown. In the present study, we observed the effects of LPS and TLR4/SIRT3 on EPCs mitochondrial permeability and intracellular mitochondrial superoxide. We employed the monocrotaline-induced pulmonary arteriolar injury model to observe the effects of TLR4/SIRT3 on the recruitment and survival of transplanted EPCs. We found the destructive effects of 10 μg/mL LPS on mitochondrial homeostasis, and cellular viability was mediated by TLR4/SIRT3 signals at least partially, and the TLR4 mediates the early-stage recruitment of transplanted EPCs in pulmonary arteriolar inflammation injury; however, SIRT3 has more contribution to the survival of incorporated EPCs and ameliorated arteriolar remodeling in lung vascular tissue. The study provides insights for the critical role of TLR4/SIRT3 in LPS-induced oxidative stress and mitochondrial disorder in EPCs in vitro and in vivo. The TLR4/SIRT3 signaling is important for EPCs resistance against inflammation and oxidative stress and may represent a new manipulating target for developing efficient cell therapy strategy.
Collapse
|
18
|
Melissa officinalis L. Supplementation Provides Cardioprotection in a Rat Model of Experimental Autoimmune Myocarditis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1344946. [PMID: 35265259 PMCID: PMC8901324 DOI: 10.1155/2022/1344946] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/09/2022] [Indexed: 12/03/2022]
Abstract
Due to existing evidence regarding antioxidant and anti-inflammatory effects of Melissa officinalis extracts (MOEs), this study was aimed at investigating the potential of ethanolic MOE to prevent the development of myocarditis and its ability to ameliorate the severity of experimental autoimmune myocarditis (EAM) by investigating MOE effects on in vivo cardiac function, structure, morphology, and oxidative stress parameters. A total of 50 7-week-old male Dark Agouti rats were enrolled in the study and randomly allocated into the following groups: CTRL, nontreated healthy rats; EAM, nontreated rats with EAM; MOE50, MOE100, and MOE200, rats with EAM treated with either 50, 100, or 200 mg/kg of MOE for 3 weeks per os. Myocarditis was induced by immunization of the rats with porcine myocardial myosin (0.5 mg) emulsion on day 0. Cardiac function and dimensions of the left ventricle (LV) were assessed via echocardiography. Additionally, the blood pressure and heart rate were measured. On day 21, rats were sacrificed and the hearts were isolated for further histopathological analyses (H/E and Picrosirius red staining). The blood samples were collected to determine oxidative stress parameters. The EAM group characteristically showed greater LV wall thickness and lower ejection fraction (50.33 ± 7.94% vs. 84.81 ± 7.74%) and fractional shortening compared to CTRL (p < 0.05). MOE significantly improved echocardiographic parameters (EF in MOE200 81.44 ± 5.51%) and also reduced inflammatory infiltrate (by 88.46%; p < 0.001) and collagen content (by 76.39%; p < 0.001) in the heart tissues, especially in the MOE200 group compared to the EAM group. In addition, MOEs induced a significant decrease of prooxidants production (O2−, H2O2, and TBARS) and improved antioxidant defense system via increase in GSH, SOD, and CAT compared to EAM, with medium and high dose being more effective than low dose (p < 0.05). The present study suggests that ethanolic MOEs, especially in a 200 mg/kg dose, improve cardiac function and myocardial architecture, possibly via oxidative stress mitigation, thus preventing heart remodeling, development of dilated cardiomyopathy, and subsequent heart failure connected with EAM. MOEs might be considered as a potentially helpful adjuvant therapy in patients with autoimmune myocarditis.
Collapse
|
19
|
Wang S, Tan J, Miao Y, Zhang Q. Mitochondrial Dynamics, Mitophagy, and Mitochondria–Endoplasmic Reticulum Contact Sites Crosstalk Under Hypoxia. Front Cell Dev Biol 2022; 10:848214. [PMID: 35281107 PMCID: PMC8914053 DOI: 10.3389/fcell.2022.848214] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/07/2022] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are double membrane organelles within eukaryotic cells, which act as cellular power houses, depending on the continuous availability of oxygen. Nevertheless, under hypoxia, metabolic disorders disturb the steady-state of mitochondrial network, which leads to dysfunction of mitochondria, producing a large amount of reactive oxygen species that cause further damage to cells. Compelling evidence suggests that the dysfunction of mitochondria under hypoxia is linked to a wide spectrum of human diseases, including obstructive sleep apnea, diabetes, cancer and cardiovascular disorders. The functional dichotomy of mitochondria instructs the necessity of a quality-control mechanism to ensure a requisite number of functional mitochondria that are present to fit cell needs. Mitochondrial dynamics plays a central role in monitoring the condition of mitochondrial quality. The fission–fusion cycle is regulated to attain a dynamic equilibrium under normal conditions, however, it is disrupted under hypoxia, resulting in mitochondrial fission and selective removal of impaired mitochondria by mitophagy. Current researches suggest that the molecular machinery underlying these well-orchestrated processes are coordinated at mitochondria–endoplasmic reticulum contact sites. Here, we establish a holistic understanding of how mitochondrial dynamics and mitophagy are regulated at mitochondria–endoplasmic reticulum contact sites under hypoxia.
Collapse
|
20
|
The mitochondrial signaling peptide MOTS-c improves myocardial performance during exercise training in rats. Sci Rep 2021; 11:20077. [PMID: 34635713 PMCID: PMC8505603 DOI: 10.1038/s41598-021-99568-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/22/2021] [Indexed: 01/01/2023] Open
Abstract
Cardiac remodeling is a physiological adaptation to aerobic exercise and which is characterized by increases in ventricular volume and the number of cardiomyocytes. The mitochondrial derived peptide MOTS-c functions as an important regulator in physical capacity and performance. Exercise elevates levels of endogenous MOTS-c in circulation and in myocardium, while MOTS-c can significantly enhance exercise capacity. However, the effects of aerobic exercise combined with MOTS-c on cardiac structure and function are unclear. We used pressure–volume conductance catheter technique to examine cardiac function in exercised rats with and without treatment with MOTS-c. Surprisingly, MOTS-c improved myocardial mechanical efficiency, enhanced cardiac systolic function, and had a tendency to improve the diastolic function. The findings suggest that using exercise supplements could be used to modulate the cardiovascular benefits of athletic training.
Collapse
|
21
|
Zhao F, Zou MH. Role of the Mitochondrial Protein Import Machinery and Protein Processing in Heart Disease. Front Cardiovasc Med 2021; 8:749756. [PMID: 34651031 PMCID: PMC8505727 DOI: 10.3389/fcvm.2021.749756] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are essential organelles for cellular energy production, metabolic homeostasis, calcium homeostasis, cell proliferation, and apoptosis. About 99% of mammalian mitochondrial proteins are encoded by the nuclear genome, synthesized as precursors in the cytosol, and imported into mitochondria by mitochondrial protein import machinery. Mitochondrial protein import systems function not only as independent units for protein translocation, but also are deeply integrated into a functional network of mitochondrial bioenergetics, protein quality control, mitochondrial dynamics and morphology, and interaction with other organelles. Mitochondrial protein import deficiency is linked to various diseases, including cardiovascular disease. In this review, we describe an emerging class of protein or genetic variations of components of the mitochondrial import machinery involved in heart disease. The major protein import pathways, including the presequence pathway (TIM23 pathway), the carrier pathway (TIM22 pathway), and the mitochondrial intermembrane space import and assembly machinery, related translocases, proteinases, and chaperones, are discussed here. This review highlights the importance of mitochondrial import machinery in heart disease, which deserves considerable attention, and further studies are urgently needed. Ultimately, this knowledge may be critical for the development of therapeutic strategies in heart disease.
Collapse
Affiliation(s)
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
22
|
D’Errico S, Russa RL, Maiese A, Santurro A, Scopetti M, Romano S, Zanon M, Frati P, Fineschi V. Atypical antipsychotics and oxidative cardiotoxicity: review of literature and future perspectives to prevent sudden cardiac death. J Geriatr Cardiol 2021; 18:663-685. [PMID: 34527032 PMCID: PMC8390928 DOI: 10.11909/j.issn.1671-5411.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress is considered the principal mediator of myocardial injury under pathological conditions. It is well known that reactive oxygen (ROS) or nitrogen species (RNS) are involved in myocardial injury and repair at the same time and that cellular damage is generally due to an unbalance between generation and elimination of the free radicals due to an inadequate mechanism of antioxidant defense or to an increase in ROS and RNS. Major adverse cardiovascular events are often associated with drugs with associated findings such as fibrosis or inflammation of the myocardium. Despite efforts in the preclinical phase of the development of drugs, cardiotoxicity still remains a great concern. Cardiac toxicity due to second-generation antipsychotics (clozapine, olanzapine, quetiapine) has been observed in preclinical studies and described in patients affected with mental disorders. A role of oxidative stress has been hypothesized but more evidence is needed to confirm a causal relationship. A better knowledge of cardiotoxicity mechanisms should address in the future to establish the right dose and length of treatment without impacting the physical health of the patients.
Collapse
Affiliation(s)
- Stefano D’Errico
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Raffaele La Russa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
| | - Aniello Maiese
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Surgical Pathology, Medical, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Santurro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Scopetti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Romano
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Martina Zanon
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Paola Frati
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Vittorio Fineschi
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
23
|
Heinen-Weiler J, Hasenberg M, Heisler M, Settelmeier S, Beerlage AL, Doepper H, Walkenfort B, Odersky A, Luedike P, Winterhager E, Rassaf T, Hendgen-Cotta UB. Superiority of focused ion beam-scanning electron microscope tomography of cardiomyocytes over standard 2D analyses highlighted by unmasking mitochondrial heterogeneity. J Cachexia Sarcopenia Muscle 2021; 12:933-954. [PMID: 34120411 PMCID: PMC8350221 DOI: 10.1002/jcsm.12742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/16/2021] [Accepted: 05/21/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cardioprotection by preventing or repairing mitochondrial damage is an unmet therapeutic need. To understand the role of cardiomyocyte mitochondria in physiopathology, the reliable characterization of the mitochondrial morphology and compartment is pivotal. Previous studies mostly relied on two-dimensional (2D) routine transmission electron microscopy (TEM), thereby neglecting the real three-dimensional (3D) mitochondrial organization. This study aimed to determine whether classical 2D TEM analysis of the cardiomyocyte ultrastructure is sufficient to comprehensively describe the mitochondrial compartment and to reflect mitochondrial number, size, dispersion, distribution, and morphology. METHODS Spatial distribution of the complex mitochondrial network and morphology, number, and size heterogeneity of cardiac mitochondria in isolated adult mouse cardiomyocytes and adult wild-type left ventricular tissues (C57BL/6) were assessed using a comparative 3D imaging system based on focused ion beam-scanning electron microscopy (FIB-SEM) nanotomography. For comparison of 2D vs. 3D data sets, analytical strategies and mathematical comparative approaches were performed. To confirm the value of 3D data for mitochondrial changes, we compared the obtained values for number, coverage area, size heterogeneity, and complexity of wild-type cardiomyocyte mitochondria with data sets from mice lacking the cytosolic and mitochondrial protein BNIP3 (BCL-2/adenovirus E1B 19-kDa interacting protein 3; Bnip3-/- ) using FIB-SEM. Mitochondrial respiration was assessed on isolated mitochondria using the Seahorse XF analyser. A cardiac biopsy was obtained from a male patient (48 years) suffering from myocarditis. RESULTS The FIB-SEM nanotomographic analysis revealed that no linear relationship exists for mitochondrial number (r = 0.02; P = 0.9511), dispersion (r = -0.03; P = 0.9188), and shape (roundness: r = 0.15, P = 0.6397; elongation: r = -0.09, P = 0.7804) between 3D and 2D results. Cumulative frequency distribution analysis showed a diverse abundance of mitochondria with different sizes in 3D and 2D. Qualitatively, 2D data could not reflect mitochondrial distribution and dynamics existing in 3D tissue. 3D analyses enabled the discovery that BNIP3 deletion resulted in more smaller, less complex cardiomyocyte mitochondria (number: P < 0.01; heterogeneity: C.V. wild-type 89% vs. Bnip3-/- 68%; complexity: P < 0.001) forming large myofibril-distorting clusters, as seen in human myocarditis with disturbed mitochondrial dynamics. Bnip3-/- mice also show a higher respiration rate (P < 0.01). CONCLUSIONS Here, we demonstrate the need of 3D analyses for the characterization of mitochondrial features in cardiac tissue samples. Hence, we observed that BNIP3 deletion physiologically acts as a molecular brake on mitochondrial number, suggesting a role in mitochondrial fusion/fission processes and thereby regulating the homeostasis of cardiac bioenergetics.
Collapse
Affiliation(s)
- Jacqueline Heinen-Weiler
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany.,Imaging Center Essen (IMCES), Electron Microscopy Unit (EMU), Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Mike Hasenberg
- Imaging Center Essen (IMCES), Electron Microscopy Unit (EMU), Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Martin Heisler
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Anna-Lena Beerlage
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Hannah Doepper
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Bernd Walkenfort
- Imaging Center Essen (IMCES), Electron Microscopy Unit (EMU), Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Andrea Odersky
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Peter Luedike
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Elke Winterhager
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany.,Imaging Center Essen (IMCES), Electron Microscopy Unit (EMU), Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
24
|
Pang Y, Zhu Z, Wen Z, Lu J, Lin H, Tang M, Xu Z, Lu J. HIGD‑1B inhibits hypoxia‑induced mitochondrial fragmentation by regulating OPA1 cleavage in cardiomyocytes. Mol Med Rep 2021; 24:549. [PMID: 34080026 PMCID: PMC8185509 DOI: 10.3892/mmr.2021.12188] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
The dynamic regulation of mitochondrial morphology is key for eukaryotic cells to manage physiological challenges. Therefore, it is important to understand the molecular basis of mitochondrial dynamic regulation. The aim of the present study was to explore the role of HIG1 hypoxia inducible domain family member 1B (HIGD‑1B) in hypoxia‑induced mitochondrial fragmentation. Protein expression was determined via western blotting. Immunofluorescence assays were performed to detect the subcellular location of HIGD‑1B. Cell Counting Kit‑8 assays and flow cytometry were carried out to measure cell viability and apoptosis, respectively. Protein interactions were evaluated by co‑immunoprecipitation. In the present study, it was found that HIGD‑1B serves a role in cell survival by maintaining the integrity of the mitochondria under hypoxic conditions. Knockdown of HIGD‑1B promoted mitochondrial fragmentation, while overexpression of HIGD‑1B increased survival by preventing activation of caspase‑3 and ‑9. HIGD‑1B expression was associated with cell viability and apoptosis in cardiomyocytes. Furthermore, HIGD‑1B delayed the cleavage process of optic atrophy 1 (OPA1) and stabilized mitochondrial morphology by interacting with OPA1. Collectively, the results from the present study identified a role for HIGD‑1B as an inhibitor of the mitochondrial fission in cardiomyocytes.
Collapse
Affiliation(s)
- Yan Pang
- Department of Cardiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R. China
| | - Zhide Zhu
- Academic Affairs Section, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Zhihao Wen
- Department of Cardiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R. China
| | - Junshen Lu
- Academic Affairs Section, Guangxi University of Traditional Chinese Medicine Attached Chinese Medicine School, Nanning, Guangxi 530001, P.R. China
| | - Hao Lin
- Department of Geriatrics, Danzhou Traditional Chinese Medicine Hospital, Danzhou, Hainan 571700, P.R. China
| | - Meiling Tang
- Department of Cardiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R. China
| | - Zhiliang Xu
- Academic Affairs Section, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Jianqi Lu
- Department of Cardiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R. China
| |
Collapse
|
25
|
Hu Y, Xu Y, Chen W, Qiu Z. Stomatin-Like Protein-2: A Potential Target to Treat Mitochondrial Cardiomyopathy. Heart Lung Circ 2021; 30:1449-1455. [PMID: 34088631 DOI: 10.1016/j.hlc.2021.05.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/16/2021] [Accepted: 05/02/2021] [Indexed: 12/27/2022]
Abstract
Stomatin-like protein-2 (SLP-2) is a mitochondrial-associated protein that is abundant in cardiomyocytes. Many reports have shown that SLP-2 plays an important role in mitochondria. The treatment of mitochondrial cardiomyopathy (MCM) needs further improvement, so the relationship between SLP-2 and MCM is worth exploring. This study reviewed some protective mechanisms of SLP-2 on mitochondria. Published studies have shown that SLP-2 protects mitochondria by stabilising the function of optic atrophy 1 (OPA1), promoting mitofusin (Mfn) 2 expression, interacting with prohibitins and cardiolipin, forming SLP-2-PARL-YME1L (SPY) complex, and stabilising respiratory chain complexes, suggesting that SLP-2 is a new potential target for the treatment of MCM. However, the specific mechanism of SLP-2 needs to be confirmed by further research.
Collapse
Affiliation(s)
- Yuntao Hu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Yueyue Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China.
| | - Zhibing Qiu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China.
| |
Collapse
|
26
|
Dowling JK, Afzal R, Gearing LJ, Cervantes-Silva MP, Annett S, Davis GM, De Santi C, Assmann N, Dettmer K, Gough DJ, Bantug GR, Hamid FI, Nally FK, Duffy CP, Gorman AL, Liddicoat AM, Lavelle EC, Hess C, Oefner PJ, Finlay DK, Davey GP, Robson T, Curtis AM, Hertzog PJ, Williams BRG, McCoy CE. Mitochondrial arginase-2 is essential for IL-10 metabolic reprogramming of inflammatory macrophages. Nat Commun 2021; 12:1460. [PMID: 33674584 PMCID: PMC7936006 DOI: 10.1038/s41467-021-21617-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Mitochondria are important regulators of macrophage polarisation. Here, we show that arginase-2 (Arg2) is a microRNA-155 (miR-155) and interleukin-10 (IL-10) regulated protein localized at the mitochondria in inflammatory macrophages, and is critical for IL-10-induced modulation of mitochondrial dynamics and oxidative respiration. Mechanistically, the catalytic activity and presence of Arg2 at the mitochondria is crucial for oxidative phosphorylation. We further show that Arg2 mediates this process by increasing the activity of complex II (succinate dehydrogenase). Moreover, Arg2 is essential for IL-10-mediated downregulation of the inflammatory mediators succinate, hypoxia inducible factor 1α (HIF-1α) and IL-1β in vitro. Accordingly, HIF-1α and IL-1β are highly expressed in an LPS-induced in vivo model of acute inflammation using Arg2-/- mice. These findings shed light on a new arm of IL-10-mediated metabolic regulation, working to resolve the inflammatory status of the cell.
Collapse
Affiliation(s)
- Jennifer K Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- FutureNeuro, SFI Research Centre, Dublin 2, Ireland
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Remsha Afzal
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Linden J Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mariana P Cervantes-Silva
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Stephanie Annett
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Gavin M Davis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Chiara De Santi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Nadine Assmann
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Daniel J Gough
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Glenn R Bantug
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Fidinny I Hamid
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Frances K Nally
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Conor P Duffy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Aoife L Gorman
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Alex M Liddicoat
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Ed C Lavelle
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Christoph Hess
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Gavin P Davey
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Annie M Curtis
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Bryan R G Williams
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Claire E McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
- FutureNeuro, SFI Research Centre, Dublin 2, Ireland.
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.
| |
Collapse
|
27
|
Weekate K, Chuenjitkuntaworn B, Chuveera P, Vaseenon S, Chompu-Inwai P, Ittichaicharoen J, Chattipakorn S, Srisuwan T. Alterations of mitochondrial dynamics, inflammation and mineralization potential of lipopolysaccharide-induced human dental pulp cells after exposure to N-acetyl cysteine, Biodentine or ProRoot MTA. Int Endod J 2021; 54:951-965. [PMID: 33503268 DOI: 10.1111/iej.13484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/25/2021] [Indexed: 12/14/2022]
Abstract
AIM To investigate the effects of N-acetyl cysteine (NAC), Biodentine, ProRoot MTA and their combinations, on cell viability, mitochondrial reactive oxygen species (mtROS) production, mineralization and on the expression of genes related to inflammatory cytokine production, mitochondrial dynamics and cell apoptosis of lipopolysaccharide (LPS)-induced human dental pulp cells (hDPCs). METHODOLOGY Isolated hDPCs were exposed to 20 μg mL-1 of Escherichia coli (E. coli) LPS for 24 h, before the experiment, except for the control group. Eight experimental groups were assigned: (i) control (hDPCs cultured in regular medium), (ii) +LPS (hDPCs cultured in LPS medium throughout the experiment), (iii) -LPS/Media, (iv) -LPS/BD, (v) -LPS/MTA, (vi) -LPS/NAC, (vii) -LPS/BD + NAC and (viii) -LPS/MTA + NAC. Cell viability was measured using Alamar blue assay at 24 and 48 h. Production of mtROS was evaluated at 6 and 24 h by MitoSOX Red and MitoTracker Green. The expressions of IL-6, TNF-α, Bcl-2, Bax, Mfn-2 and Drp-1 genes were investigated at 6 h using reverse transcriptase-polymerase chain reaction (RT-PCR). For differentiation potential, cells were cultured in the osteogenic differentiation media and stained using Alizarin red assay at 14 and 21 days. The Kruskal-Wallis test, Mann-Whitney U test and one-way anova were performed for statistical analysis. RESULTS NAC was associated with significantly greater LPS-induced hDPC viability (P < 0.05). Both Biodentine and MTA extracts promoted cell survival, whereas the combination of NAC to these material extracts significantly increased the number of viable cells at 24 h (P < 0.05). Biodentine, MTA or NAC did not alter the mtROS level (P > 0.05). NAC supplementation to the MTA extract significantly reduced the level of IL-6 and TNF-α expression (P < 0.05). Regarding mitochondrial dynamics, the use of NAC alone promoted significant Mfn-2/Drp-1 expression (P < 0.05). Most of the groups exhibited a level of Bcl-2/Bax gene expression similar to that of the control group. The increases in mineralization productions were observed in most of the groups, except the LPS group (P < 0.05). CONCLUSIONS The antioxidant effect of NAC was not evident under the LPS-induced condition in DPC in vitro. NAC combined either with Biodentine or MTA improved LPS-induced hDPCs survival at 24 h. The combination of NAC with MTA promoted mineralization.
Collapse
Affiliation(s)
- K Weekate
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - B Chuenjitkuntaworn
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - P Chuveera
- Department of Family and Community Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - S Vaseenon
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - P Chompu-Inwai
- Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - J Ittichaicharoen
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - S Chattipakorn
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - T Srisuwan
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
28
|
Sun W, Han B, Cai D, Wang J, Jiang D, Jia H. Differential Expression Profiles and Functional Prediction of Circular RNAs in Pediatric Dilated Cardiomyopathy. Front Mol Biosci 2021; 7:600170. [PMID: 33392258 PMCID: PMC7775584 DOI: 10.3389/fmolb.2020.600170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Circular RNAs (circRNAs) have emerged as essential regulators and biomarkers in various diseases. To assess the different expression levels of circRNAs in pediatric dilated cardiomyopathy (PDCM) and explore their biological and mechanistic significance, we used RNA microarrays to identify differentially expressed circRNAs between three children diagnosed with PDCM and three healthy age-matched volunteers. The biological function of circRNAs was assessed with a circRNA–microRNA (miRNA)–mRNA interaction network constructed from Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes. Differentially expressed circRNAs were validated by quantitative real-time polymerase chain reaction (qRT-PCR) in 25 children with PDCM and 25 healthy volunteers. We identified 257 up-regulated (fold change ≤ 0.5, P < 0.05) and 899 down-regulated (fold change ≥2, P < 0.05) circRNAs in PDCM patients when compared to healthy volunteers. The qRT-PCR experiments confirmed has_circ_0067735 down-regulation (0.45-fold, P < 0.001), has_circ_0070186 up-regulation (2.82-fold, P < 0.001), and has_circ_0069972 down-regulation (0.50-fold, P < 0.05). A functional analysis of these differentially expressed circRNAs suggests that they are associated with hypertrophy, remodeling, fibrosis, and autoimmunity. CircRNAs have been implicated in PDCM through largely unknown mechanisms. Here we report differentially expressed circRNAs in PDCM patients that may illuminate the mechanistic roles in the etiology of PDCM that could serve as non-invasive diagnostic biomarkers.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongxiao Cai
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Wang
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Diandong Jiang
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hailin Jia
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
29
|
Katare PB, Nizami HL, Paramesha B, Dinda AK, Banerjee SK. Activation of toll like receptor 4 (TLR4) promotes cardiomyocyte apoptosis through SIRT2 dependent p53 deacetylation. Sci Rep 2020; 10:19232. [PMID: 33159115 PMCID: PMC7648754 DOI: 10.1038/s41598-020-75301-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/13/2020] [Indexed: 12/31/2022] Open
Abstract
Cardiomyocyte inflammation followed by apoptosis and fibrosis is an important mediator for development and progression of heart failure. Activation of toll-like receptor 4 (TLR4), an important regulator of inflammation, causes the progression of cardiac hypertrophy and injury. However, the precise mechanism of TLR4-mediated adverse cardiac outcomes is still elusive. The present study was designed to find the role of TLR4 in cardiac fibrosis and apoptosis, and molecular mechanism thereof. Rats were treated with TLR4 agonist (LPS 12.5 μg/kg/day) through osmotic pump for 14 days. To simulate the condition in vitro, H9c2 cells were treated with LPS (1 μg/ml). Similarly, H9c2 cells were transfected with TLR4 and SIRT2 c-DNA clone for overexpression. Myocardial oxidative stress, inflammation, fibrosis and mitochondrial parameters were evaluated both in vitro and in vivo. Cardiac inflammation after LPS treatment was confirmed by increased TNF-α and IL-6 expression in rat heart. There was a marked increase in oxidative stress as observed by increased TBARS and decreased endogenous antioxidants (GSH and catalase), along with mitochondrial dysfunction as measured by mitochondrial complex activity in LPS-treated rat hearts. Histopathological examination showed the presence of cardiac fibrosis after LPS treatment. Protein expression of nuclear p53 and cleaved caspase-7/caspase-9 was significantly increased in LPS treated heart. Similar to in vivo study, nuclear translocation of p53, mitochondrial dysfunction and cellular apoptosis were observed in H9c2 cells treated with LPS. Our data also indicate that decreased expression of SIRT2 was associated with increased acetylation of p53 after LPS treatment. In conclusion, TLR4 activation in rats promotes cardiac inflammation, mitochondrial dysfunction, apoptosis and fibrosis. p53 and caspase 7/caspase 9 were found to play an important role in TLR4-mediated apoptosis. Our data suggest that, reducing TLR4 mediated fibrosis and apoptosis could be a novel approach in the treatment of heart failure, keeping in the view the major role played by TLR4 in cardiac inflammation.
Collapse
Affiliation(s)
- Parmeshwar Bajirao Katare
- Drug Discovery Research Centre (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Hina Lateef Nizami
- Drug Discovery Research Centre (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Bugga Paramesha
- Drug Discovery Research Centre (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Amit K Dinda
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029, India
| | - Sanjay K Banerjee
- Drug Discovery Research Centre (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India.
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India.
| |
Collapse
|
30
|
Hernandez‐Resendiz S, Prunier F, Girao H, Dorn G, Hausenloy DJ, EU‐CARDIOPROTECTION COST Action (CA16225). Targeting mitochondrial fusion and fission proteins for cardioprotection. J Cell Mol Med 2020; 24:6571-6585. [PMID: 32406208 PMCID: PMC7299693 DOI: 10.1111/jcmm.15384] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 01/05/2023] Open
Abstract
New treatments are needed to protect the myocardium against the detrimental effects of acute ischaemia/reperfusion (IR) injury following an acute myocardial infarction (AMI), in order to limit myocardial infarct (MI) size, preserve cardiac function and prevent the onset of heart failure (HF). Given the critical role of mitochondria in energy production for cardiac contractile function, prevention of mitochondrial dysfunction during acute myocardial IRI may provide novel cardioprotective strategies. In this regard, the mitochondrial fusion and fissions proteins, which regulate changes in mitochondrial morphology, are known to impact on mitochondrial quality control by modulating mitochondrial biogenesis, mitophagy and the mitochondrial unfolded protein response. In this article, we review how targeting these inter-related processes may provide novel treatment targets and new therapeutic strategies for reducing MI size, preventing the onset of HF following AMI.
Collapse
Affiliation(s)
- Sauri Hernandez‐Resendiz
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Centro de Biotecnologia‐FEMSATecnologico de MonterreyNuevo LeonMexico
| | - Fabrice Prunier
- Institut MITOVASCCNRS UMR 6015 INSERM U1083University Hospital Center of AngersUniversity of AngersAngersFrance
| | - Henrique Girao
- Faculty of MedicineCoimbra Institute for Clinical and Biomedical Research (iCBR)University of CoimbraPortugal
- Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Clinical Academic Centre of Coimbra (CACC)CoimbraPortugal
| | - Gerald Dorn
- Department of Internal MedicineCenter for PharmacogenomicsWashington University School of MedicineSt. LouisMOUSA
| | - Derek J. Hausenloy
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular Research CenterCollege of Medical and Health SciencesAsia UniversityTaichungTaiwan
| | | |
Collapse
|
31
|
Kespohl M, Bredow C, Klingel K, Voß M, Paeschke A, Zickler M, Poller W, Kaya Z, Eckstein J, Fechner H, Spranger J, Fähling M, Wirth EK, Radoshevich L, Thery F, Impens F, Berndt N, Knobeloch KP, Beling A. Protein modification with ISG15 blocks coxsackievirus pathology by antiviral and metabolic reprogramming. SCIENCE ADVANCES 2020; 6:eaay1109. [PMID: 32195343 PMCID: PMC7065878 DOI: 10.1126/sciadv.aay1109] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/13/2019] [Indexed: 05/10/2023]
Abstract
Protein modification with ISG15 (ISGylation) represents a major type I IFN-induced antimicrobial system. Common mechanisms of action and species-specific aspects of ISGylation, however, are still ill defined and controversial. We used a multiphasic coxsackievirus B3 (CV) infection model with a first wave resulting in hepatic injury of the liver, followed by a second wave culminating in cardiac damage. This study shows that ISGylation sets nonhematopoietic cells into a resistant state, being indispensable for CV control, which is accomplished by synergistic activity of ISG15 on antiviral IFIT1/3 proteins. Concurrent with altered energy demands, ISG15 also adapts liver metabolism during infection. Shotgun proteomics, in combination with metabolic network modeling, revealed that ISG15 increases the oxidative capacity and promotes gluconeogenesis in liver cells. Cells lacking the activity of the ISG15-specific protease USP18 exhibit increased resistance to clinically relevant CV strains, therefore suggesting that stabilizing ISGylation by inhibiting USP18 could be exploited for CV-associated human pathologies.
Collapse
Affiliation(s)
- Meike Kespohl
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Germany
| | - Clara Bredow
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | - Karin Klingel
- University of Tuebingen, Cardiopathology, Institute for Pathology and Neuropathology, Tuebingen, Germany
| | - Martin Voß
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | - Anna Paeschke
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | - Martin Zickler
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | - Wolfgang Poller
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Clinic for Cardiology, Campus Benjamin Franklin, Berlin, Germany
| | - Ziya Kaya
- Universitätsklinikum Heidelberg, Medizinische Klinik für Innere Medizin III: Kardiologie, Angiologie und Pneumologie, Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Heidelberg, Germany
| | - Johannes Eckstein
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | - Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Joachim Spranger
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Michael Fähling
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Vegetative Physiology, Berlin, Germany
| | - Eva Katrin Wirth
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Fabien Thery
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Center for Medical Biotechnology, Ghent, Belgium
- VIB Proteomics Core, Ghent, Belgium
| | - Nikolaus Berndt
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute for Computational and Imaging Science in Cardiovascular Medicine, Berlin, Germany
| | | | - Antje Beling
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Germany
- Corresponding author.
| |
Collapse
|
32
|
Jin B, Shi H, Zhu J, Wu B, Geshang Q. Up-regulating autophagy by targeting the mTOR-4EBP1 pathway: a possible mechanism for improving cardiac function in mice with experimental dilated cardiomyopathy. BMC Cardiovasc Disord 2020; 20:56. [PMID: 32019530 PMCID: PMC6998347 DOI: 10.1186/s12872-020-01365-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/29/2020] [Indexed: 01/22/2023] Open
Abstract
Background Autophagy plays a crucial role in the pathological process of cardiovascular diseases. However, little is known about the pathological mechanism underlying autophagy regulation in dilated cardiomyopathy (DCM). Methods We explored whether up-regulating autophagy could improve cardiac function in mice with experimental DCM through the mTOR-4EBP1 pathway. Animal model of DCM was established in BALB/c mice by immunization with porcine cardiac myosin. Both up- or down-regulation of autophagy were studied by administration of rapamycin or 3-MA in parallel. Morphology, Western blotting, and echocardiography were applied to confirm the pathological mechanisms. Results Autophagy was activated and autophagosomes were significantly increased in the rapamycin group. The collagen volume fraction (CVF) was decreased in the rapamycin group compared with the DCM group (9.21 ± 0.82% vs 14.38 ± 1.24%, P < 0.01). The expression of p-mTOR and p-4EBP1 were significantly decreased in rapamycin-induced autophagy activation, while the levels were increased by down-regulating autophagy with 3-MA. In the rapamycin group, the LVEF and FS were significantly increased compared with the DCM group (54.12 ± 6.48% vs 45.29 ± 6.68%, P < 0.01; 26.89 ± 4.04% vs 22.17 ± 2.82%, P < 0.05). As the inhibitor of autophagy, 3-MA aggravated the progress of maladaptive cardiac remodeling and declined cardiac function in DCM mice. Conclusions The study indicated a possible mechanism for improving cardiac function in mice with experimental DCM by up-regulating autophagy via the mTOR-4EBP1 pathway, which could be a promising therapeutic strategy for DCM.
Collapse
Affiliation(s)
- Bo Jin
- Department of Cardiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Haiming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jun Zhu
- Department of Cardiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Bangwei Wu
- Department of Cardiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Quzhen Geshang
- Department of Medicine, Medical College of Tibet University, Lasa, Tibet, China
| |
Collapse
|
33
|
Sacchetto C, Sequeira V, Bertero E, Dudek J, Maack C, Calore M. Metabolic Alterations in Inherited Cardiomyopathies. J Clin Med 2019; 8:E2195. [PMID: 31842377 PMCID: PMC6947282 DOI: 10.3390/jcm8122195] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
The normal function of the heart relies on a series of complex metabolic processes orchestrating the proper generation and use of energy. In this context, mitochondria serve a crucial role as a platform for energy transduction by supplying ATP to the varying demand of cardiomyocytes, involving an intricate network of pathways regulating the metabolic flux of substrates. The failure of these processes results in structural and functional deficiencies of the cardiac muscle, including inherited cardiomyopathies. These genetic diseases are characterized by cardiac structural and functional anomalies in the absence of abnormal conditions that can explain the observed myocardial abnormality, and are frequently associated with heart failure. Since their original description, major advances have been achieved in the genetic and phenotype knowledge, highlighting the involvement of metabolic abnormalities in their pathogenesis. This review provides a brief overview of the role of mitochondria in the energy metabolism in the heart and focuses on metabolic abnormalities, mitochondrial dysfunction, and storage diseases associated with inherited cardiomyopathies.
Collapse
Affiliation(s)
- Claudia Sacchetto
- IMAiA—Institute for Molecular Biology and RNA Technology, Faculty of Health, Universiteitssingel 50, 6229ER Maastricht, The Netherlands;
- Medicine and Life Sciences, Faculty of Science and Engineering, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
- Department of Biology, University of Padova, via Ugo Bassi 58B, 35121 Padova, Italy
| | - Vasco Sequeira
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Edoardo Bertero
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Jan Dudek
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Christoph Maack
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 9708 Würzburg, Germany; (V.S.); (E.B.); (J.D.)
| | - Martina Calore
- IMAiA—Institute for Molecular Biology and RNA Technology, Faculty of Health, Universiteitssingel 50, 6229ER Maastricht, The Netherlands;
- Medicine and Life Sciences, Faculty of Science and Engineering, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| |
Collapse
|
34
|
Li S, Wang Y, Zhao C, Zhang M, Wang W, Yu X, Huang J, Wang Z, Zhu B, Yin C, Cai H. Akt inhibitor deguelin aggravates inflammation and fibrosis in myocarditis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1275-1282. [PMID: 32128091 PMCID: PMC7038425 DOI: 10.22038/ijbms.2019.35518.8473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 05/12/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Myocarditis is characterized by inflammatory cell infiltration in myocardial stroma. Attenuation of tumor necrosis factor (TNF)-α and interleukin (IL)-1β is a reliable mark for improving the prognosis. Protein kinase B (Akt) plays an important role in the development and progression of myocarditis. The specific role of the natural inhibitor of Akt, Deguelin, on myocarditis has not been reported. In this study, we used deguelin to investigate the effects of natural Akt inhibitor on myocarditis in experimental autoimmune myocarditis (EAM) rats. MATERIALS AND METHODS EAM rat models were made by using Lewis rats and Deguelin was injected intraperitoneally on day 3, 6, 9, 12 and 15 after successful modeling. On day 18, rats were sacrificed and the heart weight (HW)/ body weight (BW) ratio were measured. The pathological changes, pathological scores and fibrosis area were evaluated after H.&E. and Masson's trichrome staining. The mRNA levels of TNF-α and IL-1β were measured by RT-qPCR, while the protein expressions of TNF-α and IL-1β were detected by immunohistochemical staining and Western bolt. The protein expressions of Akt, Akt1, phosphorylated (p-) Akt and nuclear factor (NF)-κB were detected by Western bolt. RESULTS We found that the TNF-α and IL-1β levels, inflammatory scores and fibrosis areas were markedly increased after 18 days deguelin administration. CONCLUSION Akt inhibition with deguelin may aggravate myocarditis of EAM rats.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yue Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chunming Zhao
- Human anatomy and Histology and Embryology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Meixiang Zhang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaowei Yu
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jiao Huang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhao Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Bo Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Chengqian Yin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Hongxing Cai
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
35
|
Fu CY, Chen J, Lu XY, Zheng MZ, Wang LL, Shen YL, Chen YY. Dimethyl fumarate attenuates lipopolysaccharide-induced mitochondrial injury by activating Nrf2 pathway in cardiomyocytes. Life Sci 2019; 235:116863. [PMID: 31513817 DOI: 10.1016/j.lfs.2019.116863] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
AIMS To determine whether dimethyl fumarate (DMF) can protect against lipopolysaccharide (LPS) -induced myocardial injury. MAIN METHODS H9c2 cells pretreated with or without DMF were stimulated with LPS. Cell viability and apoptosis were evaluated. Nrf2 and HO-1 expression were detected using Western blotting. Mitochondrial morphology, mitochondrial superoxide production were observed using confocal microscope. Mitochondrial respiration function was measured using Seahorse bioanalyzer. KEY FINDINGS (1) The cell viability decreased, LDH release and apoptosis increased in LPS- challenged H9c2 cells. DMF pretreatment brought a higher cell viability, and a lower LDH leakage and apoptosis than those of LPS group (P < 0.01). (2) DMF pretreatment resulted in an increased Nrf2 and HO-1 expression, and enhanced nuclear Nrf2 level in LPS-challenged cells (P < 0.01). (3) Nrf2-siRNA could inhibit DMF-induced enhancement of HO-1 expression and cell viability, and partly abolish DMF-induced reduction of LDH leakage and apoptosis. (4) ERK1/2 inhibitor PD98059 could not only prevent the DMF-induced enhancement of nuclear Nrf2 and HO-1, but also inhibit DMF-induced increase in cell viability. (5) Compared with LPS-challenged cells, DMF pretreatment caused a lower production of mitochondrial superoxide and a higher mitochondrial membrane potential, which could be abolished by Nrf2-siRNA. (6) DMF could attenuate LPS-induced mitochondrial fragmentation and improve mitochondrial respiration function by enhancement of the oxygen consumption rate of basal respiration and ATP production in LPS-challenged cells (P < 0.01). SIGNIFICANCE DMF protects cardiomyocytes against LPS-induced damage. ERK1/2-dependent activation of Nrf2/HO-1 pathway is responsible for DMF-induced cardioprotection via reduction of oxidative stress, improvement of mitochondrial morphology and energy metabolism.
Collapse
Affiliation(s)
- Chun-Yan Fu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jun Chen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Yang Lu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ming-Zhi Zheng
- Department of Pharmacology, Hangzhou Medical College, Hangzhou 310053, China
| | - Lin-Lin Wang
- Center for Stem Cell and Tissue Engineering, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yue-Liang Shen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Ying-Ying Chen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
36
|
Exogenous Hydrogen Sulfide Supplement Attenuates Isoproterenol-Induced Myocardial Hypertrophy in a Sirtuin 3-Dependent Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9396089. [PMID: 30647820 PMCID: PMC6311776 DOI: 10.1155/2018/9396089] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/11/2018] [Indexed: 01/25/2023]
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter with a variety of cardiovascular protective effects. Sirtuin 3 (SIRT3) is closely related to mitochondrial function and oxidative stress. We found that NaHS increased SIRT3 expression in the preventive effect on isoproterenol- (ISO-) induced myocardial hypertrophy. We further investigated whether exogenous H2S supplement improved ISO-induced myocardial hypertrophy in a SIRT3-dependent manner. 10-week-old male 129S1/SvImJ (WT) mice and SIRT3 knockout (KO) mice were intraperitoneally injected with NaHS (50 μmol/kg/d) for two weeks and then intraperitoneally injected with ISO (60 mg/kg/d) for another two weeks. In WT mice, NaHS significantly reduced the cardiac index of ISO-induced mice, decreased the cross-sectional area of cardiomyocytes, and inhibited the expressions of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) mRNA. The activity of total antioxidant capacity (T-AOC) and superoxide dismutase (SOD) in the myocardium was increased, but the level of malondialdehyde (MDA) was decreased. The fluorescence intensity of dihydroethidium staining for superoxide anion was attenuated. Optic atrophy 1 (OPA1) expression was upregulated, while dynamin-related protein 1 (DRP1) expression was downregulated. ERK, but not P38 and JNK, phosphorylation was downregulated. However, all above protective effects were unavailable in ISO-induced SIRT3 KO mice. Our present study suggested that exogenous H2S supplement inhibited ISO-induced cardiac hypertrophy depending on SIRT3, which might be associated with antioxidant stress.
Collapse
|
37
|
Jin B, Zhu J, Shi HM, Wen ZC, Wu BW. YAP activation promotes the transdifferentiation of cardiac fibroblasts to myofibroblasts in matrix remodeling of dilated cardiomyopathy. ACTA ACUST UNITED AC 2018; 52:e7914. [PMID: 30484494 PMCID: PMC6262745 DOI: 10.1590/1414-431x20187914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/27/2018] [Indexed: 11/29/2022]
Abstract
Yes-associated protein (YAP) is an important regulator of cellular proliferation and transdifferentiation. However, little is known about the mechanisms underlying myofibroblast transdifferentiation in dilated cardiomyopathy (DCM). We investigated the role of YAP in the pathological process of cardiac matrix remodeling. A classic model of DCM was established in BALB/c mice by immunization with porcine cardiac myosin. Cardiac fibroblasts were isolated from neonatal Sprague-Dawley rats by density gradient centrifugation. The expression levels of α-smooth muscle actin (α-SMA) and collagen volume fraction (CVF) were significantly increased in DCM mice. Angiotensin II (Ang II)-mediated YAP activation promoted the proliferation and transdifferentiation of neonatal rat cardiac fibroblasts, and this effect was significantly suppressed in the shRNA YAP + Ang II group compared with the shRNA Control + Ang II group in vitro (2.98±0.34 ×105vs 5.52±0.82 ×105, P<0.01). Inhibition of endogenous Ang II-stimulated YAP improved the cardiac function by targeting myofibroblast transdifferentiation to attenuate matrix remodeling in vivo. In the valsartan group, left ventricular ejection fraction and fractional shortening were significantly increased compared with the DCM group (52.72±5.51% vs 44.46±3.01%, P<0.05; 34.84±3.85% vs 26.65±3.12%, P<0.01). Our study demonstrated that YAP was a regulator of cardiac myofibroblast differentiation, and regulation of YAP signaling pathway contributed to improve cardiac function of DCM mice, possibly in part by decreasing myofibroblast transdifferentiation to inhibit matrix remodeling.
Collapse
Affiliation(s)
- Bo Jin
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Zhu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hai-Ming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhi-Chao Wen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bang-Wei Wu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
38
|
A Disturbance in the Force: Cellular Stress Sensing by the Mitochondrial Network. Antioxidants (Basel) 2018; 7:antiox7100126. [PMID: 30249006 PMCID: PMC6211095 DOI: 10.3390/antiox7100126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/13/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
As a highly dynamic organellar network, mitochondria are maintained as an organellar network by delicately balancing fission and fusion pathways. This homeostatic balance of organellar dynamics is increasingly revealed to play an integral role in sensing cellular stress stimuli. Mitochondrial fission/fusion balance is highly sensitive to perturbations such as loss of bioenergetic function, oxidative stress, and other stimuli, with mechanistic contribution to subsequent cell-wide cascades including inflammation, autophagy, and apoptosis. The overlapping activity with m-AAA protease 1 (OMA1) metallopeptidase, a stress-sensitive modulator of mitochondrial fusion, and dynamin-related protein 1 (DRP1), a regulator of mitochondrial fission, are key factors that shape mitochondrial dynamics in response to various stimuli. As such, OMA1 and DRP1 are critical factors that mediate mitochondrial roles in cellular stress-response signaling. Here, we explore the current understanding and emerging questions in the role of mitochondrial dynamics in sensing cellular stress as a dynamic, responsive organellar network.
Collapse
|