1
|
Borkum JM. Cluster Headache and Hypoxia: Breathing New Life into an Old Theory, with Novel Implications. Neurol Int 2024; 16:1691-1716. [PMID: 39728749 DOI: 10.3390/neurolint16060123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Cluster headache is a severe, poorly understood disorder for which there are as yet virtually no rationally derived treatments. Here, Lee Kudrow's 1983 theory, that cluster headache is an overly zealous response to hypoxia, is updated according to current understandings of hypoxia detection, signaling, and sensitization. It is shown that the distinctive clinical characteristics of cluster headache (circadian timing of attacks and circannual patterning of bouts, autonomic symptoms, and agitation), risk factors (cigarette smoking; male gender), triggers (alcohol; nitroglycerin), genetic findings (GWAS studies), anatomical substrate (paraventricular nucleus of the hypothalamus, solitary tract nucleus/NTS, and trigeminal nucleus caudalis), neurochemical features (elevated levels of galectin-3, nitric oxide, tyramine, and tryptamine), and responsiveness to treatments (verapamil, lithium, melatonin, prednisone, oxygen, and histamine desensitization) can all be understood in terms of hypoxic signaling. Novel treatment directions are hypothesized, including repurposing pharmacological antagonists of hypoxic signaling molecules (HIF-2; P2X3) for cluster headache, breath training, physical exercise, high-dose thiamine, carnosine, and the flavonoid kaempferol. The limits of current knowledge are described, and a program of basic and translational research is proposed.
Collapse
Affiliation(s)
- Jonathan M Borkum
- Department of Psychology, University of Maine, 301 Williams Hall, Orono, ME 04469-5742, USA
| |
Collapse
|
2
|
Skarp S, Doedens A, Holmström L, Izzi V, Saarimäki S, Sliz E, Kettunen J, Pakanen L, Kerkelä R, Pylkäs K, Huikuri HV, Myerburg RJ, Junttila J. Novel Genetic Variants Associated with Primary Myocardial Fibrosis in Sudden Cardiac Death Victims. J Cardiovasc Transl Res 2024; 17:1229-1239. [PMID: 38848015 PMCID: PMC11634914 DOI: 10.1007/s12265-024-10527-5] [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: 01/23/2024] [Accepted: 05/21/2024] [Indexed: 12/13/2024]
Abstract
Myocardial fibrosis is a common finding in victims of sudden cardiac death (SCD). Whole exome sequencing was performed in 127 victims of SCD with primary myocardial fibrosis as the only pathological finding. These cases are derived from the Fingesture study which has collected data from autopsy-verified SCD victims in Northern Finland. A computational approach was used to identify protein interactions in cardiomyocytes. Associations of the identified variants with cardiac disease endpoints were investigated in the Finnish national genetic study (FinnGen) dataset. We identified 21 missense and one nonsense variant. Four variants were estimated to affect protein function, significantly associated with SCD/primary myocardial fibrosis (Fingesture) and associated with cardiac diseases in Finnish population (FinnGen). These variants locate in cartilage acidic protein 1 (CRATC1), calpain 1 (CAPN1), unc-45 myosin chaperone A (UNC45A) and unc-45 myosin chaperone B (UNC45B). The variants identified contribute to function of extracellular matrix and cardiomyocytes.
Collapse
Affiliation(s)
- Sini Skarp
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | - Anne Doedens
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Lauri Holmström
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Valerio Izzi
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Samu Saarimäki
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Eeva Sliz
- Systems Medicine, Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Johannes Kettunen
- Systems Medicine, Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Lasse Pakanen
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, Oulu, Finland
| | - Risto Kerkelä
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Katri Pylkäs
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Heikki V Huikuri
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Robert J Myerburg
- Division of Cardiology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juhani Junttila
- Research unit of Biomedicine and Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Biocenter Oulu, Faculty of Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
3
|
Zhang X, Zheng Y, Wang Z, Zhang G, Yang L, Gan J, Jiang X. Calpain: The regulatory point of cardiovascular and cerebrovascular diseases. Biomed Pharmacother 2024; 179:117272. [PMID: 39153432 DOI: 10.1016/j.biopha.2024.117272] [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: 05/12/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Calpain, a key member of the Calpain cysteine protease superfamily, performs limited protein hydrolysis in a calcium-dependent manner. Its activity is tightly regulated due to the potential for non-specific cleavage of various intracellular proteins upon aberrant activation. A thorough review of the literature from 2010 to 2023 reveals 121 references discussing cardiovascular and cerebrovascular diseases. Dysregulation of the Calpain system is associated with various pathological phenomena, including lipid metabolism disorders, inflammation, apoptosis, and excitotoxicity. Although recent studies have revealed the significant role of Calpain in cardiovascular and cerebrovascular diseases, the precise mechanisms remain incompletely understood. Exploring the potential of Calpain inhibition as a therapeutic approach for the treatment of cardiovascular and cerebrovascular diseases may emerge as a compelling area of interest for future calpain research.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yujia Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Guangming Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
4
|
Onal M, Elsurer C, Duran T, Kocak N, Ulusoy B, Bozkurt MK, Onal O. Possible role of endoplasmic reticulum stress in the pathogenesis of chronic adenoiditis and adenoid hypertrophy: A prospective, parallel-group study. Laryngoscope Investig Otolaryngol 2024; 9:e1240. [PMID: 38596230 PMCID: PMC11002993 DOI: 10.1002/lio2.1240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Background Adenoid tissue is a first-line host defense secondary lymphoid organ, especially in childhood. The endoplasmic reticulum (ER) is required to maintain balanced cellular activity. With impaired ER functions, protein accumulation occurs, resulting in ER stress, which plays a role in the etiopathogenesis of many diseases. Objective We aimed to investigate the relationship between ER stress and adenoid tissue disorders, thereby elucidating the mechanisms of immunity-related diseases. Methods Fifty-four pediatric patients (>3 years old) who underwent adenoidectomy for chronic adenoiditis (CA) or adenoid hypertrophy (AH) were enrolled in this prospective, parallel-group clinical study. Adenoids were divided into two groups (CA or AH) based on their size and evaluated for ER stress pathway and apoptosis pathway markers by Real-time PCR and Western blot analysis. Results ER stress pathway markers significantly differed between the CA and AH groups. Children with CA had higher ER stress marker levels than the AH group (p < .001 for ATF-4, ATF-6, and GRP78, and p < .05 for EDEM1, CHOP, EIF2AK3, ERNI, and GRP94). Apoptosis pathway marker levels (BAX and BCL-2) were not different between groups. Conclusions ER stress contributes to the etiopathogenesis of adenoid tissue diseases and the pathogenesis of adenoid tissue disorders, which are part of the immune response. These results may guide the development of new and alternative treatments for immune system disorders.
Collapse
Affiliation(s)
- Merih Onal
- Department of OtorhinolaryngologySelcuk University Faculty of MedicineKonyaTurkey
| | - Cagdas Elsurer
- Department of OtorhinolaryngologySelcuk University Faculty of MedicineKonyaTurkey
| | - Tugce Duran
- Department of Medical GeneticsKTO Karatay University Faculty of MedicineKonyaTurkey
| | - Nadir Kocak
- Department of Medical GeneticsSelcuk University Faculty of MedicineKonyaTurkey
| | - Bulent Ulusoy
- Department of OtorhinolaryngologySelcuk University Faculty of MedicineKonyaTurkey
| | - Mete Kaan Bozkurt
- Department of OtorhinolaryngologySelcuk University Faculty of MedicineKonyaTurkey
| | - Ozkan Onal
- Department of Anesthesiology and ReanimationSelcuk University Faculty of MedicineKonyaTurkey
- Outcomes Research ConsortiumCleveland Clinic Main Hospital, Anesthesiology InstituteClevelandOhioUSA
| |
Collapse
|
5
|
Song SE, Shin SK, Ju HY, Im SS, Song DK. Role of cytosolic and endoplasmic reticulum Ca 2+ in pancreatic beta-cells: pros and cons. Pflugers Arch 2024; 476:151-161. [PMID: 37940681 DOI: 10.1007/s00424-023-02872-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
Pancreatic beta cells utilize Ca2+ to secrete insulin in response to glucose. The glucose-dependent increase in cytosolic Ca2+ concentration ([Ca2+]C) activates a series of insulin secretory machinery in pancreatic beta cells. Therefore, the amount of insulin secreted in response to glucose is determined in a [Ca2+]C-dependent manner, at least within a moderate range. However, the demand for insulin secretion may surpass the capability of beta cells. Abnormal elevation of [Ca2+]C levels beyond the beta-cell endurance capacity can damage them by inducing endoplasmic reticulum (ER) stress and cell death programs such as apoptosis. Therefore, while Ca2+ is essential for the insulin secretory functions of beta cells, it could affect their survival at pathologically higher levels. Because an increase in beta-cell [Ca2+]C is inevitable under certain hazardous conditions, understanding the regulatory mechanism for [Ca2+]C is important. Therefore, this review discusses beta-cell function, survival, ER stress, and apoptosis associated with intracellular and ER Ca2+ homeostasis.
Collapse
Affiliation(s)
- Seung-Eun Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, South Korea
| | - Hyeon Yeong Ju
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung-Soon Im
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Dae-Kyu Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea.
| |
Collapse
|
6
|
Wang Y, Li M, Chen J, Yu Y, Yu Y, Shi H, Liu X, Chen Z, Chen R, Ge J. Macrophage CAPN4 regulates CVB3-induced cardiac inflammation and injury by promoting NLRP3 inflammasome activation and phenotypic transformation to the inflammatory subtype. Free Radic Biol Med 2023; 208:430-444. [PMID: 37660839 DOI: 10.1016/j.freeradbiomed.2023.08.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Exploring the immune mechanism of coxsackievirus B3 (CVB3)-induced myocarditis may provide a promising therapeutic strategy. Here, we investigated the regulatory role of macrophage CAPN4 in the phenotypic transformation of macrophages and NOD-like receptor protein 3 (NLRP3) inflammasome activation. We found that CAPN4 was the most upregulated subtype of the calpain family in CVB3-infected bone marrow-derived macrophages (BMDMs) and Raw 264.7 cells after CVB3 infection and was upregulated in cardiac macrophages from CVB3-infected mice. Conditional knockout of CAPN4 (CAPN4flox/flox; LYZ2-Cre, CAPN4-cKO mice) ameliorated inflammation and myocardial injury and improved cardiac function and survival after CVB3 infection. Enrichment analysis revealed that macrophage differentiation and the interleukin signaling pathway were the most predominant biological processes in macrophages after CVB3 infection. We further found that CVB3 infection and the overexpression of CAPN4 promoted macrophage M1 polarization and NLRP3 inflammasome activation, while CAPN4 knockdown reversed these changes. Correspondingly, CAPN4-cKO alleviated CVB3-induced M1 macrophage transformation and NLRP3 expression and moderately increased M2 transformation in vivo. The culture supernatant of CAPN4-overexpressing or CVB3-infected macrophages impaired cardiac fibroblast function and viability. Moreover, macrophage CAPN4 could upregulate C/EBP-homologous protein (chop) expression, which increased proinflammatory cytokine release by activating the phosphorylation of transducer of activator of transcription 1 (STAT1) and 3 (STAT3). Overall, these results suggest that CAPN4 increases M1-type and inhibits M2-type macrophage polarization through the chop-STAT1/STAT3 signaling pathway to mediate CVB3-induced myocardial inflammation and injury. CAPN4 may be a novel target for viral myocarditis treatment.
Collapse
Affiliation(s)
- Yucheng Wang
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Minghui Li
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Jun Chen
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Ying Yu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Yong Yu
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Hui Shi
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Xiaoxiao Liu
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Zhiwei Chen
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Ruizhen Chen
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China.
| | - Junbo Ge
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| |
Collapse
|
7
|
Xue X, Xi W, Li W, Xiao J, Wang Z, Zhang Y. Hydrogen-rich saline alleviates cardiomyocyte apoptosis by reducing expression of calpain1 via miR-124-3p. ESC Heart Fail 2023; 10:3077-3090. [PMID: 37602925 PMCID: PMC10567641 DOI: 10.1002/ehf2.14492] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 06/16/2023] [Accepted: 07/16/2023] [Indexed: 08/22/2023] Open
Abstract
AIMS Molecular hydrogen has been exhibited a protective function in heart diseases. Our previous study demonstrated that hydrogen-rich saline (HRS) could scavenge free radicals selectively and alleviate the inflammatory response in the myocardial ischaemia/reperfusion (I/R) injury, but the underlying mechanism has not been fully clarified. METHODS AND RESULTS Adult (10 weeks) C57BL/6 male mice and neonatal rat cardiomyocytes were used to establish I/R and hypoxia/reoxygenation (H/R) injury models. I/R and H/R models were treated with HRS to classify the mechanisms of cardioproctective function. In this study, we found that miR-124-3p was significantly decreased in both I/R and H/R models, while it was partially ameliorated by HRS pretreatment. HRS treatment also alleviated ischaemia-induced apoptotic cell death and increased cell viability during I/R process, whereas silencing expression of miR-124-3p abolished this protective effect. In addition, we identified calpain1 as a direct target of miR-124-3p, and up-regulation of miR-124-3 produced both activity and expression of calpain1. It was also found that compared with the HRS group, overexpression of calpain1 increased caspase-3 activities, promoted cleaved-caspase3 and Bax protein expressions, and correspondingly decreased Bcl-2, further reducing cell viability. These results illustrated that calpain1 overexpression attenuated protective effect of HRS on cardiomyocytes in H/R model. CONCLUSIONS The present study showed a protective effect of HRS on I/R injury, which may be associated with miR-124-3p-calpain1 signalling pathway.
Collapse
Affiliation(s)
- Xiaofei Xue
- Department of Cardiothoracic SurgeryChangzheng Hospital, Naval Military Medical University415 Fengyang RoadShanghai200003China
| | - Wang Xi
- Department of Cardiothoracic SurgeryChangzheng Hospital, Naval Military Medical University415 Fengyang RoadShanghai200003China
| | - Wei Li
- Department of Cardiothoracic SurgeryGeneral Hospital of Central Theater CommandWuhanChina
| | - Jian Xiao
- Department of Cardiothoracic SurgeryChangzheng Hospital, Naval Military Medical University415 Fengyang RoadShanghai200003China
| | - Zhinong Wang
- Department of Cardiothoracic SurgeryChangzheng Hospital, Naval Military Medical University415 Fengyang RoadShanghai200003China
| | - Yufeng Zhang
- Department of Cardiothoracic SurgeryChangzheng Hospital, Naval Military Medical University415 Fengyang RoadShanghai200003China
| |
Collapse
|
8
|
Zhang RR, Zhang JL, Li Q, Zhang SM, Gu XM, Niu W, Zhou JJ, Zhou LC. SEVERE BURN-INDUCED MITOCHONDRIAL RECRUITMENT OF CALPAIN CAUSES ABERRANT MITOCHONDRIAL DYNAMICS AND HEART DYSFUNCTION. Shock 2023; 60:255-261. [PMID: 37278996 PMCID: PMC10476594 DOI: 10.1097/shk.0000000000002159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/12/2023] [Accepted: 05/31/2023] [Indexed: 06/07/2023]
Abstract
ABSTRACT Mitochondrial damage is an important cause of heart dysfunction after severe burn injury. However, the pathophysiological process remains unclear. This study aims to examine the mitochondrial dynamics in the heart and the role of μ-calpain, a cysteine protease, in this scenario. Rats were subjected to severe burn injury treatment, and the calpain inhibitor MDL28170 was administered intravenously 1 h before or after burn injury. Rats in the burn group displayed weakened heart performance and decreased mean arterial pressure, which was accompanied by a diminishment of mitochondrial function. The animals also exhibited higher levels of calpain in mitochondria, as reflected by immunofluorescence staining and activity tests. In contrast, treatment with MDL28170 before any severe burn diminished these responses to a severe burn. Burn injury decreased the abundance of mitochondria and resulted in a lower percentage of small mitochondria and a higher percentage of large mitochondria. Furthermore, burn injury caused an increase in the fission protein DRP1 in the mitochondria and a decrease in the inner membrane fusion protein OPA1. Similarly, these alterations were also blocked by MDL28170. Of note, inhibition of calpain yielded the emergence of more elongated mitochondria along with membrane invagination in the middle of the longitude, which is an indicator of the fission process. Finally, MDL28170, administered 1 h after burn injury, preserved mitochondrial function and heart performance, and increased the survival rate. Overall, these results provided the first evidence that mitochondrial recruitment of calpain confers heart dysfunction after severe burn injury, which involves aberrant mitochondrial dynamics.
Collapse
Affiliation(s)
- Ran-Ran Zhang
- School of Life Science, Northwest University, Xi’an, China
| | - Jing-Long Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Xi’an, China
| | - Qiao Li
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Xi’an, China
| | - Shu-Miao Zhang
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi’an, China
| | - Xiao-Ming Gu
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi’an, China
| | - Wen Niu
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi’an, China
| | - Jing-Jun Zhou
- School of Life Science, Northwest University, Xi’an, China
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi’an, China
| | - Lyu-Chen Zhou
- School of Life Science, Northwest University, Xi’an, China
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi’an, China
| |
Collapse
|
9
|
Liu GY, Xie WL, Wang YT, Chen L, Xu ZZ, Lv Y, Wu QP. Calpain: the regulatory point of myocardial ischemia-reperfusion injury. Front Cardiovasc Med 2023; 10:1194402. [PMID: 37456811 PMCID: PMC10346867 DOI: 10.3389/fcvm.2023.1194402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Calpain is a conserved cysteine protease readily expressed in several mammalian tissues, which is usually activated by Ca2+ and with maximum activity at neutral pH. The activity of calpain is tightly regulated because its aberrant activation will nonspecifically cleave various proteins in cells. Abnormally elevation of Ca2+ promotes the abnormal activation of calpain during myocardial ischemia-reperfusion, resulting in myocardial injury and cardiac dysfunction. In this paper, we mainly reviewed the effects of calpain in various programmed cell death (such as apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, and parthanatos) in myocardial ischemia-reperfusion. In addition, we also discussed the abnormal activation of calpain during myocardial ischemia-reperfusion, the effect of calpain on myocardial repair, and the possible future research directions of calpain.
Collapse
Affiliation(s)
- Guo-Yang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Wan-Li Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yan-Ting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhen-Zhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yong Lv
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Qing-Ping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| |
Collapse
|
10
|
Abdel-Megeed RM, Kadry MO. Amelioration of autophagy and inflammatory signaling pathways via α-lipoic acid, burdock and bee pollen versus lipopolysaccharide-induced insulin resistance in murine model. Heliyon 2023; 9:e15692. [PMID: 37139293 PMCID: PMC10149403 DOI: 10.1016/j.heliyon.2023.e15692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 05/05/2023] Open
Abstract
Lipopolysaccharide (LPS) has previously been implicated in insulin resistance by generating an innate immune response and activating inflammatory cascades. Many studies have discovered a relationship between high levels of serum LPS and the advancement of diabetic microvascular problems, indicating that LPS may play a role in the control of critical signaling pathways connected to insulin resistance. The current study focused on signaling pathways linked to insulin resistance and explored probable mechanisms of LPS-induced insulin resistance in a murine model. It next looked at the effects of burdock, bee pollen, and -lipoic acid on LPS-induced inflammation and autoimmune defects in rats. LPS intoxication was induced via ip injection for one week in a dose of 10 mg/kg followed by α-lipoic acid, Burdock and bee pollen in an oral treatment for one month. Following that, biochemical and molecular studies were performed. The RNA expression of the regulating genes STAT5A and PTEN was measured. In addition, ATF-4 and CHOP as autophagy biomarkers were also subjected to mRNA quantification. The results demonstrated a considerable improvement in the -lipoic acid, Burdock, and bee pollen treated groups via modifying oxidative stress indicators as well as molecular ones. Furthermore, glucose concentration in serum and α-amylase were also improved upon treatment with the superiority of α-lipoic acid for modulating all estimated parameters. In conclusion: the results declared in the current study suggested that α-lipoic acid could regulate insulin resistance signaling pathways induced by LPS intoxication.
Collapse
|
11
|
Ji XY, Zheng D, Ni R, Wang JX, Shao JQ, Vue Z, Hinton A, Song LS, Fan GC, Chakrabarti S, Su ZL, Peng TQ. Sustained over-expression of calpain-2 induces age-dependent dilated cardiomyopathy in mice through aberrant autophagy. Acta Pharmacol Sin 2022; 43:2873-2884. [PMID: 35986214 PMCID: PMC9622835 DOI: 10.1038/s41401-022-00965-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 07/24/2022] [Indexed: 11/09/2022] Open
Abstract
Calpains have been implicated in heart diseases. While calpain-1 has been detrimental to the heart, the role of calpain-2 in cardiac pathology remains controversial. In this study we investigated whether sustained over-expression of calpain-2 had any adverse effects on the heart and the underlying mechanisms. Double transgenic mice (Tg-Capn2/tTA) were generated, which express human CAPN2 restricted to cardiomyocytes. The mice were subjected to echocardiography at age 3, 6, 8 and 12 months, and their heart tissues and sera were collected for analyses. We showed that transgenic mice over-expressing calpain-2 restricted to cardiomyocytes had normal heart function with no evidence of cardiac pathological remodeling at age 3 months. However, they exhibited features of dilated cardiomyopathy including increased heart size, enlarged heart chambers and heart dysfunction from age 8 months; histological analysis revealed loss of cardiomyocytes replaced by myocardial fibrosis and cardiomyocyte hypertrophy in transgenic mice from age 8 months. These cardiac alterations closely correlated with aberrant autophagy evidenced by significantly increased LC3BII and p62 protein levels and accumulation of autophagosomes in the hearts of transgenic mice. Notably, injection of 3-methyladenine, a well-established inhibitor of autophagy (30 mg/kg, i.p. once every 3 days starting from age 6 months for 2 months) prevented aberrant autophagy, attenuated myocardial injury and improved heart function in the transgenic mice. In cultured cardiomyocytes, over-expression of calpain-2 blocked autophagic flux by impairing lysosomal function. Furthermore, over-expression of calpain-2 resulted in lower levels of junctophilin-2 protein in the heart of transgenic mice and in cultured cardiomyocytes, which was attenuated by 3-methyladenine. In addition, blockade of autophagic flux by bafilomycin A (100 nM) induced a reduction of junctophilin-2 protein in cardiomyocytes. In summary, transgenic over-expression of calpain-2 induces age-dependent dilated cardiomyopathy in mice, which may be mediated through aberrant autophagy and a reduction of junctophilin-2. Thus, a sustained increase in calpain-2 may be detrimental to the heart.
Collapse
Affiliation(s)
- Xiao-Yun Ji
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada
| | - Dong Zheng
- Centre of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Rui Ni
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada
| | - Jin-Xi Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jian-Qiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada
| | - Zhao-Liang Su
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China.
| | - Tian-Qing Peng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada.
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada.
- Department of Medicine, Western University, London, ON, N6A 5W9, Canada.
| |
Collapse
|
12
|
Tunicamycin-Induced Endoplasmic Reticulum Stress Damages Complex I in Cardiac Mitochondria. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081209. [PMID: 36013387 PMCID: PMC9409705 DOI: 10.3390/life12081209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Induction of acute ER (endoplasmic reticulum) stress using thapsigargin contributes to complex I damage in mouse hearts. Thapsigargin impairs complex I by increasing mitochondrial calcium through inhibition of Ca2+-ATPase in the ER. Tunicamycin (TUNI) is used to induce ER stress by inhibiting protein folding. We asked if TUNI-induced ER stress led to complex I damage. METHODS TUNI (0.4 mg/kg) was used to induce ER stress in C57BL/6 mice. Cardiac mitochondria were isolated after 24 or 72 h following TUNI treatment for mitochondrial functional analysis. RESULTS ER stress was only increased in mice following 72 h of TUNI treatment. TUNI treatment decreased oxidative phosphorylation with complex I substrates compared to vehicle with a decrease in complex I activity. The contents of complex I subunits including NBUPL and NDUFS7 were decreased in TUNI-treated mice. TUNI treatment activated both cytosolic and mitochondrial calpain 1. Our results indicate that TUNI-induced ER stress damages complex I through degradation of its subunits including NDUFS7. CONCLUSION Induction of the ER stress using TUNI contributes to complex I damage by activating calpain 1.
Collapse
|
13
|
Perner C, Krüger E. Endoplasmic Reticulum Stress and Its Role in Homeostasis and Immunity of Central and Peripheral Neurons. Front Immunol 2022; 13:859703. [PMID: 35572517 PMCID: PMC9092946 DOI: 10.3389/fimmu.2022.859703] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Neuronal cells are specialists for rapid transfer and translation of information. Their electrical properties relay on a precise regulation of ion levels while their communication via neurotransmitters and neuropeptides depends on a high protein and lipid turnover. The endoplasmic Reticulum (ER) is fundamental to provide these necessary requirements for optimal neuronal function. Accumulation of misfolded proteins in the ER lumen, reactive oxygen species and exogenous stimulants like infections, chemical irritants and mechanical harm can induce ER stress, often followed by an ER stress response to reinstate cellular homeostasis. Imbedded between glial-, endothelial-, stromal-, and immune cells neurons are constantly in communication and influenced by their local environment. In this review, we discuss concepts of tissue homeostasis and innate immunity in the central and peripheral nervous system with a focus on its influence on ER stress, the unfolded protein response, and implications for health and disease.
Collapse
Affiliation(s)
- Caroline Perner
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Neurology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
14
|
Chen J, Liu Y, Pan D, Xu T, Luo Y, Wu W, Wu P, Zhu H, Li D. Estrogen inhibits endoplasmic reticulum stress and ameliorates myocardial ischemia/reperfusion injury in rats by upregulating SERCA2a. Cell Commun Signal 2022; 20:38. [PMID: 35331264 PMCID: PMC8944077 DOI: 10.1186/s12964-022-00842-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background The incidence of coronary heart disease (CHD) in premenopausal women is significantly lower than that of men of the same age, suggesting protective roles of estrogen for the cardiovascular system against CHD. This study aimed to confirm the protective effect of estrogen on myocardium during myocardial ischemia/reperfusion (MI/R) injury and explore the underlying mechanisms. Methods Neonatal rat cardiomyocytes and Sprague–Dawley rats were used in this study. Different groups were treated by bilateral ovariectomy, 17β-estradiol (E2), adenoviral infection, or siRNA transfection. The expression of sarcoplasmic reticulum Ca2+ ATPase pump (SERCA2a) and endoplasmic reticulum (ER) stress-related proteins were measured in each group to examine the effect of different E2 levels and determine the relationship between SERCA2a and ER stress. The cell apoptosis, myocardial infarction size, levels of apoptosis and serum cardiac troponin I, ejection fraction, calcium transient, and morphology changes of the myocardium and ER were examined to verify the effects of E2 on the myocardium. Results Bilateral ovariectomy resulted in reduced SERCA2a levels and more severe MI/R injury. E2 treatment increased SERCA2a expression. Both E2 treatment and exogenous SERCA2a overexpression decreased levels of ER stress-related proteins and alleviated myocardial damage. In contrast, SERCA2a knockdown exacerbated ER stress and myocardial damage. Addition of E2 after SERCA2a knockdown did not effectively inhibit ER stress or reduce myocardial injury. Conclusions Our data demonstrate that estrogen inhibits ER stress and attenuates MI/R injury by upregulating SERCA2a. These results provide a new potential target for therapeutic intervention and drug discovery in CHD. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00842-2.
Collapse
Affiliation(s)
- Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Tongda Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Chen Q, Thompson J, Hu Y, Lesnefsky EJ. Reversing mitochondrial defects in aged hearts: role of mitochondrial calpain activation. Am J Physiol Cell Physiol 2022; 322:C296-C310. [PMID: 35044856 PMCID: PMC8836732 DOI: 10.1152/ajpcell.00279.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
Aging chronically increases endoplasmic reticulum (ER) stress that contributes to mitochondrial dysfunction. Activation of calpain 1 (CPN1) impairs mitochondrial function during acute ER stress. We proposed that aging-induced ER stress led to mitochondrial dysfunction by activating CPN1. We posit that attenuation of the ER stress or direct inhibition of CPN1 in aged hearts can decrease cardiac injury during ischemia-reperfusion by improving mitochondrial function. Male young (3 mo) and aged mice (24 mo) were used in the present study, and 4-phenylbutyrate (4-PBA) was used to decrease the ER stress in aged mice. Subsarcolemmal (SSM) and interfibrillar mitochondria (IFM) were isolated. Chronic 4-PBA treatment for 2 wk decreased CPN1 activation as shown by the decreased cleavage of spectrin in cytosol and apoptosis inducing factor (AIF) and the α1 subunit of pyruvate dehydrogenase (PDH) in mitochondria. Treatment improved oxidative phosphorylation in 24-mo-old SSM and IFM at baseline compared with vehicle. When 4-PBA-treated 24-mo-old hearts were subjected to ischemia-reperfusion, infarct size was decreased. These results support that attenuation of the ER stress decreased cardiac injury in aged hearts by improving mitochondrial function before ischemia. To challenge the role of CPN1 as an effector of the ER stress, aged mice were treated with MDL-28170 (MDL, an inhibitor of calpain 1). MDL treatment improved mitochondrial function in aged SSM and IFM. MDL-treated 24-mo-old hearts sustained less cardiac injury following ischemia-reperfusion. These results support that age-induced ER stress augments cardiac injury during ischemia-reperfusion by impairing mitochondrial function through activation of CPN1.
Collapse
Affiliation(s)
- Qun Chen
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jeremy Thompson
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ying Hu
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
- McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| |
Collapse
|
16
|
Impairment of μ-calpain activation by rhTNFR:Fc reduces severe burn-induced membrane disruption in the heart. Cell Death Dis 2022; 8:10. [PMID: 35013173 PMCID: PMC8748603 DOI: 10.1038/s41420-021-00810-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 11/08/2022]
Abstract
Stress cardiomyopathy is a major clinical complication after severe burn. Multiple upstream initiators have been identified; however, the downstream targets are not fully understood. This study assessed the role of the plasma membrane in this process and its relationship with the protease μ-calpain and tumor necrosis factor-alpha (TNF-α). Here, third-degree burn injury of approximately 40% of the total body surface area was established in rats. Plasma levels of LDH and cTnI and cardiac cell apoptosis increased at 0.5 h post burn, reached a peak at 6 h, and gradually declined at 24 h. This effect correlated well with not only the disruption of cytoskeletal proteins, including dystrophin and ankyrin-B, but also with the activation of μ-calpain, as indicated by the cleaved fragments of α-spectrin and membrane recruitment of the catalytic subunit CAPN1. More importantly, these alterations were diminished by blocking calpain activity with MDL28170. Burn injury markedly increased the cellular uptake of Evans blue, indicating membrane integrity disruption, and this effect was also reversed by MDL28170. Compared with those in the control group, cardiac cells in the burn plasma-treated group were more prone to damage, as indicated by a marked decrease in cell viability and increases in LDH release and apoptosis. Of note, these alterations were mitigated by CAPN1 siRNA. Moreover, after neutralizing TNF-α with rhTNFR:Fc, calpain activity was blocked, and heart function was improved. In conclusion, we identified μ-calpain as a trigger for severe burn-induced membrane disruption in the heart and provided evidence for the application of rhTNFR:Fc to inhibit calpain for cardioprotection.
Collapse
|
17
|
Li L, Thompson J, Hu Y, Lesnefsky EJ, Willard B, Chen Q. Calpain-mediated protein targets in cardiac mitochondria following ischemia-reperfusion. Sci Rep 2022; 12:138. [PMID: 34997008 PMCID: PMC8741987 DOI: 10.1038/s41598-021-03947-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Calpain 1 and 2 (CPN1/2) are calcium-dependent cysteine proteases that exist in cytosol and mitochondria. Pharmacologic inhibition of CPN1/2 decreases cardiac injury during ischemia (ISC)-reperfusion (REP) by improving mitochondrial function. However, the protein targets of CPN1/2 activation during ISC-REP are unclear. CPN1/2 include a large subunit and a small regulatory subunit 1 (CPNS1). Genetic deletion of CPNS1 eliminates the activities of both CPN1 and CPN2. Conditional cardiomyocyte specific CPNS1 deletion mice were used in the present study to clarify the role of CPN1/2 activation in mitochondrial damage during ISC-REP with an emphasis on identifying the potential protein targets of CPN1/2. Isolated hearts from wild type (WT) or CPNS1 deletion mice underwent 25 min in vitro global ISC and 30 min REP. Deletion of CPNS1 led to decreased cytosolic and mitochondrial calpain 1 activation compared to WT. Cardiac injury was decreased in CPNS1 deletion mice following ISC-REP as shown by the decreased infarct size compared to WT. Compared to WT, mitochondrial function was improved in CPNS1 deletion mice following ischemia-reperfusion as shown by the improved oxidative phosphorylation and decreased susceptibility to mitochondrial permeability transition pore opening. H2O2 generation was also decreased in mitochondria from deletion mice following ISC-REP compared to WT. Deletion of CPNS1 also resulted in less cytochrome c and truncated apoptosis inducing factor (tAIF) release from mitochondria. Proteomic analysis of the isolated mitochondria showed that deletion of CPNS1 increased the content of proteins functioning in regulation of mitochondrial calcium homeostasis (paraplegin and sarcalumenin) and complex III activity. These results suggest that activation of CPN1 increases cardiac injury during ischemia-reperfusion by impairing mitochondrial function and triggering cytochrome c and tAIF release from mitochondria into cytosol.
Collapse
Affiliation(s)
- Ling Li
- Proteomics Core, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jeremy Thompson
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Ying Hu
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, 23298, USA
- McGuire Department of Veterans Affairs Medical Center, Richmond, VA, 23249, USA
| | - Belinda Willard
- Proteomics Core, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Qun Chen
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
18
|
Chinese Herbal Medicine Alleviates Myocardial Ischemia/Reperfusion Injury by Regulating Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4963346. [PMID: 34917158 PMCID: PMC8670943 DOI: 10.1155/2021/4963346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia/reperfusion injury is the main cause of increased mortality and disability in cardiovascular diseases. The injury involves many pathological processes, such as oxidative stress, calcium homeostasis imbalance, inflammation, and energy metabolism disorders, and these pathological stimuli can activate endoplasmic reticulum stress. In the early stage of ischemia, endoplasmic reticulum stress alleviates the injury as an adaptive survival response, but the long-term stress on endoplasmic reticulum amplifies oxidative stress, inflammation, and calcium overload to accelerate cell damage and apoptosis. Therefore, regulation of endoplasmic reticulum stress may be a mechanism to improve ischemia/reperfusion injury. Chinese herbal medicine has a long history of clinical application and unique advantages in the treatment of ischemic heart diseases. This review focuses on the effect of Chinese herbal medicine on myocardial ischemia/reperfusion injury from the perspective of regulation of endoplasmic reticulum stress.
Collapse
|
19
|
Shi H, Yu Y, Wang Y, Liu X, Yu Y, Li M, Zou Y, Chen R, Ge J. Inhibition of Calpain Alleviates Apoptosis in Coxsackievirus B3-induced Acute Virus Myocarditis Through Suppressing Endoplasmic Reticulum Stress. Int Heart J 2021; 62:900-909. [PMID: 34234076 DOI: 10.1536/ihj.20-803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Virus myocarditis (VMC) is a common cardiovascular disease and a major cause of sudden death in young adults. However, there is still a lack of effective treatments. Our previous studies found that calpain activation was involved in VMC pathogenesis. This study aims to explore the underlying mechanisms further. Neonatal rat cardiomyocytes (NRCMs) and transgenic mice overexpressing calpastatin (Tg-CAST), the endogenous calpain inhibitor, were used to establish VMC model. Hematoxylin and eosin and Masson staining revealed inflammatory cell infiltration and fibrosis. An ELISA array detected myocardial injury. Cardiac function was measured using echocardiography. CVB3 replication was assessed by capsid protein VP1. Apoptosis was measured by TUNEL staining, flow cytometry, and western blot. The endoplasmic reticulum (ER) stress-related proteins were detected by western blot. Our data showed that CVB3 infection resulted in cardiac injury, as evidenced by increased inflammatory responses and fibrosis, which induced myocardial apoptosis. Inhibiting calpain, both by PD150606 and calpastatin overexpression, could attenuate these effects. Furthermore, ER stress was activated during CVB3 infection. However, calpain inhibition could downregulate some ER stress-associated protein levels such as GRP78, pancreatic ER kinase-like ER kinase (PERK), and inositol-requiring enzyme-1α (IRE-1α), and ER stress-related apoptotic factors, during CVB3 infection. In conclusion, calpain inhibition attenuated CVB3-induced myocarditis by suppressing ER stress, thereby inhibiting cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Hui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Ying Yu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Yucheng Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Xiaoxiao Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Yong Yu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Minghui Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Ruizhen Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University
| |
Collapse
|
20
|
Zhang X, Deng R, Zhang S, Deng J, Jia JJ, Sun B, Zhou X, Bai J. Thioredoxin-1 regulates calcium homeostasis in MPP + /MPTP-induced Parkinson's disease models. Eur J Neurosci 2021; 54:4827-4837. [PMID: 34132424 DOI: 10.1111/ejn.15355] [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: 02/26/2021] [Revised: 06/04/2021] [Accepted: 06/12/2021] [Indexed: 11/30/2022]
Abstract
Disturbance in calcium (Ca2+ ) homeostasis has been involved in a variety of neuropathological conditions including Parkinson's disease (PD). The Ca2+ channel, transient receptor potential channel 1 (TRPC1), plays a protective role in regulating entry of Ca2+ activated by store depletion of Ca2+ in endoplasmic reticulum (ER). We have showed that thioredoxin-1 (Trx-1) plays a role in suppressing ER stress in PD. However, whether Trx-1 regulates TRPC1 expression in PD is still unknown. In the present study, we demonstrated that treatment of 1-methyl-4-phenylpyridinum ion (MPP+ ) significantly reduced the expression of TRPC1 in PC12 cells, which was restored by Trx-1 overexpression, and further decreased significantly by Trx-1 siRNA. Moreover, we found that Ca2+ entered into the cells was decreased by MPP+ in PC 12 cells, which was restored by Trx-1 overexpression, and further decreased by Trx-1 siRNA. MPP+ significantly increased calcium-dependent cysteine protease calpain1 expression in PC12 cells, which was suppressed by Trx-1 overexpression. Calpain1 expression was increased by Trx-1 siRNA or SKF96365, an inhibitor of TRPC1. Moreover, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) decreased TRPC1 expression in the substantia nigra pars compacta region (SNpc), which was restored in mice overexpressing Trx-1, and further decreased in mice of knockdown Trx-1. Inversely, the expression of calpain1 was increased by MPTP, which was suppressed in mice overexpressing Trx-1, and further increased in mice of knockdown Trx-1. In conclusion, Trx-1 regulates the Ca2+ entry through regulating TRPC1 expression after treatment of MPP+ /MPTP.
Collapse
Affiliation(s)
- Xianwen Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Ruhua Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Se Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Juan Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jing Jing Jia
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Bo Sun
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Xiaoshuang Zhou
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
21
|
Zheng D, Cao T, Zhang LL, Fan GC, Qiu J, Peng TQ. Targeted inhibition of calpain in mitochondria alleviates oxidative stress-induced myocardial injury. Acta Pharmacol Sin 2021; 42:909-920. [PMID: 32968209 PMCID: PMC8149722 DOI: 10.1038/s41401-020-00526-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022] Open
Abstract
The protein levels and activities of calpain-1 and calpain-2 are increased in cardiac mitochondria under pathological conditions including ischemia, diabetes, and sepsis, and transgenic overexpression of mitochondrial-targeted calpain-1 induces dilated heart failure, which underscores an important role of increased calpain in mitochondria in mediating myocardial injury. However, it remains to be determined whether selective inhibition of calpain in mitochondria protects the heart under pathological conditions. In this study, we generated transgenic mice overexpressing mitochondrial-targeted calpastatin in cardiomyocytes. Their hearts were isolated and subjected to global ischemia/reperfusion. Hyperglycemia was induced in the transgenic mice by injections of STZ. We showed that transgenic calpastatin was expressed exclusively in mitochondria isolated from their hearts but not from other organs including skeletal muscle and lung tissues. Transgenic overexpression of mitochondrial-targeted calpastatin significantly attenuated mitochondrial oxidative stress and cell death induced by global ischemia/reperfusion in isolated hearts, and ameliorated mitochondrial oxidative stress, cell death, myocardial remodeling and dysfunction in STZ-treated transgenic mice. The protective effects of mitochondrial-targeted calpastatin were correlated with increased ATP5A1 protein expression and ATP synthase activity in isolated hearts subjected to global ischemia/reperfusion and hearts of STZ-treated transgenic mice. In cultured rat myoblast H9c2 cells, overexpression of mitochondrial-targeted calpastatin maintained the protein levels of ATP5A1 and ATP synthase activity, prevented mitochondrial ROS production and decreased cell death following hypoxia/reoxygenation, whereas upregulation of ATP5A1 or scavenging of mitochondrial ROS by mito-TEMPO abrogated mitochondrial ROS production and decreased cell death. These results confirm the role of calpain in myocardial injury, suggesting that selective inhibition of calpain in myocardial mitochondria by mitochondrial-targeted calpastatin is an effective strategy for alleviating myocardial injury and dysfunction in cardiac pathologies.
Collapse
Affiliation(s)
- Dong Zheng
- Centre of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ting Cao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lu-Lu Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jun Qiu
- Centre of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Tian-Qing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
22
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
23
|
Ma D, Suh DH, Zhang J, Chao Y, Duttlinger AW, Johnson JS, Lee CH, Kim YHB. Elucidating the involvement of apoptosis in postmortem proteolysis in porcine muscles from two production cycles using metabolomics approach. Sci Rep 2021; 11:3465. [PMID: 33568769 PMCID: PMC7876139 DOI: 10.1038/s41598-021-82929-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/27/2021] [Indexed: 11/13/2022] Open
Abstract
Apoptosis has been suggested as the first step in the process of conversion of muscle into meat. While a potential role of apoptosis in postmortem proteolysis has been proposed, the underlying mechanisms by which metabolome changes in muscles would influence apoptotic and proteolytic process, leading to meat quality variation, has not been determined. Here, apoptotic and proteolytic attributes and metabolomics profiling of longissimus dorsi (LD) and psoas major (PM) muscles in pigs from two different production cycles (July–Jan vs. Apr–Sep) were evaluated. PM showed higher mitochondrial membrane permeability (MMP), concurrent with less extent of calpain-1 autolysis and troponin T degradation and higher abundance of HSP27 and αβ-crystallin compared to LD (P < 0.05). Apr–Sep muscles showed concurrence of extended apoptosis (indicated by higher MMP), calpain-1 autolysis and troponin T degradation, regardless of muscle effects (P < 0.05). Metabolomics profiling showed Apr–Sep muscles to increase in oxidative stress-related macronutrients, including 6-carbon sugars, some branched-chain AA, and free fatty acids. Antioxidant AA (His and Asp) and ascorbic acid were higher in July–Jan (P < 0.05). The results of the present study suggest that early postmortem apoptosis might be positively associated with pro-oxidant macronutrients and negatively associated with antioxidant metabolites, consequently affecting meat quality attributes in a muscle-specific manner.
Collapse
Affiliation(s)
- Danyi Ma
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Dong Ho Suh
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea
| | - Jiaying Zhang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Yufan Chao
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Alan W Duttlinger
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.,USDA-ARS Livestock Behavior Research Unit, West Lafayette, IN, 47907, USA
| | - Jay S Johnson
- USDA-ARS Livestock Behavior Research Unit, West Lafayette, IN, 47907, USA
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea. .,Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul, 05029, South Korea.
| | - Yuan H Brad Kim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
24
|
Ruan Y, Zeng J, Jin Q, Chu M, Ji K, Wang Z, Li L. Endoplasmic reticulum stress serves an important role in cardiac ischemia/reperfusion injury (Review). Exp Ther Med 2020; 20:268. [PMID: 33199993 PMCID: PMC7664614 DOI: 10.3892/etm.2020.9398] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Although acute myocardial infarction is one of the most common fatal diseases worldwide, the understanding of its underlying pathogenesis continues to develop. Myocardial ischemia/reperfusion (I/R) can restore myocardial oxygen and nutrient supply. However, a large number of studies have demonstrated that recovery of blood perfusion after acute ischemia causes reperfusion injury to the heart. With progress made in the understanding of the underlying mechanisms of myocardial I/R and oxidative stress, a novel area of research that merits greater study has been identified, that of I/R-induced endoplasmic reticulum (ER) stress (ERS). Cardiac I/R can alter the function of the ER, leading to the accumulation of unfolded/misfolded proteins. The resulting ERS then induces the activation of signal transduction pathways, which in turn contribute to the development of I/R injury. The mechanism of I/R injury, and the causal relationship between I/R and ERS are reviewed in the present article.
Collapse
Affiliation(s)
- Yongxue Ruan
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Jingjing Zeng
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Qike Jin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Maoping Chu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Kangting Ji
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Zhongyu Wang
- Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Lei Li
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
25
|
Sun F, Du J, Li H, Hao S, Zhao G, Lu F. FABP4 inhibitor BMS309403 protects against hypoxia-induced H9c2 cardiomyocyte apoptosis through attenuating endoplasmic reticulum stress. J Cell Mol Med 2020; 24:11188-11197. [PMID: 32896039 PMCID: PMC7576298 DOI: 10.1111/jcmm.15666] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/11/2020] [Accepted: 07/03/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myocardial infarction is characterized by ischaemia-induced cardiomyocyte apoptosis, in which the endoplasmic reticulum (ER) stress plays an important role. The fatty acid-binding protein-4 (FABP4) has been implicated in regulating ER stress and apoptosis. Yet, whether FABP4 is involved in modulating cardiomyocyte apoptosis remains unclarified. By applying an in vitro model of hypoxia-induced apoptosis of H9c2 cardiomyocytes, we found that FABP4 expression was elevated upon hypoxia stimulation, which was further demonstrated to be transcriptionally activated by the hypoxia-inducible factor 1a (HIF-1α). In addition, the pharmacological inhibition of FABP4 with BMS309403 protected against hypoxia-induced apoptosis in cardiomyocytes, indicating that FABP4 induction is detrimental for cardiomyocyte survival under hypoxic condition. Moreover, BMS309403 attenuated ER stress in cardiomyocytes exposed to hypoxia, which, however, was reversed by tunicamycin, an ER stress activator. More importantly, the protective effect of BMS309403 on cardiomyocytes vanished in the presence of tunicamycin. Thus, these observations establish that FABP4 inhibitor BMS309403 reduces hypoxia-induced cardiomyocyte apoptosis through attenuating excessive ER stress, implying that FABP4 inhibition may be of clinical benefit for MI treatment.
Collapse
Affiliation(s)
- Fuqiang Sun
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiangchuan Du
- Department of UltrasoundThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hongbin Li
- Department of Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shuang Hao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guochang Zhao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fanfan Lu
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
26
|
An updated role of astragaloside IV in heart failure. Biomed Pharmacother 2020; 126:110012. [DOI: 10.1016/j.biopha.2020.110012] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
|
27
|
CaMKII/calpain interaction mediates ischemia/reperfusion injury in isolated rat hearts. Cell Death Dis 2020; 11:388. [PMID: 32439852 PMCID: PMC7242471 DOI: 10.1038/s41419-020-2605-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 01/05/2023]
Abstract
Previous studies indicated that Ca2+/calmodulin-dependent kinase II (CaMKII), a kinase involved in the modulation of ryanodine receptor activity, activates Ca2+-regulated protease μ-calpain to promote myocardial ischemia/reperfusion injury. This study was performed to explore the underlying mechanisms in CaMKII-induced calpain activation to better understand heart injury. To examine the Ca2+ paradox and ischemia/reperfusion injury, isolated rat hearts were subjected to a Ca2+-free solution for 3 min, or left coronary artery occlusion for 40 min, prior to restoration of normal perfusion. Blockade of trans-sarcoplasmic reticulum Ca2+ flux using ryanodine and thapsigargin failed to prevent Ca2+ paradox-induced heart injury. In contrast, the Ca2+ paradox increased CaMKII auto-phosphorylation at Thr287, while the CaMKII inhibitor KN-62 and the Na+/Ca2+ exchanger inhibitor KB-R7943 alleviated heart injury and calpain activity. Intriguingly, the binding of μ-calpain large subunit calpain-1 (CAPN1) to phospho-CaMKII was blunted by both inhibitors. Thus, a Ca2+ leak via the ryanodine receptor is not an essential element in CaMKII-elicited calpain activation. In hearts receiving vector injection, ischemia/reperfusion caused elevated calpain activity and α-fodrin degradation, along with membrane integrity damage, similar to the effects noted in control hearts. Importantly, all these alterations were diminished with delivery of adeno-associated virus expressing mutant CaMKIIδC T287A. Ischemia/reperfusion increased CaMKII auto-phosphorylation and binding of CAPN1 to phospho-CaMKII, and facilitated the translocation of phospho-CaMKII and CAPN1 to the plasma membrane, all of which were reversed by injecting CaMKII mutant. Furthermore, the relocation capacity and the interaction of CaMKII with CAPN1 appeared to be dependent upon CaMKII autophosphorylation, as its mutant delivery increased the level of CaMKII, but did not increase membrane content of CaMKII and CAPN1, or their interactions. Together, CaMKII/calpain interaction represents a new avenue for mediating myocardial ischemia/reperfusion injury, and CaMKII likely serves as both a kinase and a carrier, thereby promoting calpain membrane translocation and activation.
Collapse
|
28
|
Taurine Prevented Hypoxia Induced Chicken Cardiomyocyte Apoptosis Through the Inhibition of Mitochondrial Pathway Activated by Calpain-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31468422 DOI: 10.1007/978-981-13-8023-5_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Objective To determine whether taurine has protective effects on chicken myocardial apoptosis induced by hypoxic condition through inhibiting calpain-1 derived mitochondrial apoptotic pathway. Methods Chicken primary embryonic myocardial cells were isolated and cultured at 37 °C under a 5% CO2 atmosphere. Firstly the optimum concentration of taurine or PD150606 was chosen by detecting the cell viability. Chicken cardiomyocytes were cultured in 95% N2-5% CO2 atmosphere for 12 h to produce hypoxic conditions. Before hypoxic treatment, 10 mM taurine and 10 uM PD150606 (a specific calpains inhibitor) were added separately or together. The cell apoptosis was detected by acridine orange/ethidium bromide (AO/EB) double staining. Western blotting was used to determine the protein expressions of calpain-1, cytochrome c, Bcl-2, procaspase-9 and procaspase-3 in the cardiomyocytes. Results Taurine administration effectively attenuated the myocardial apoptosis under hypoxic condition, reduced the calpain-1 protein level. In addition, pre-treated taurine could up-regulate the protein expressions of Bcl-2 and procaspase-3 in hypoxic myocardial cells, down-regulate protein expression levels of cytochrome c and procaspase-9. Moreover, taurine exhibited same inhibition effect as PD150606 on the cell apoptosis and proteins express under hypoxic condition. Conclusions Taurine could attenuate the chicken cardiomyocyte apoptosis impaired by hypoxia through inhibiting calpian-1-derived mitochondrial apoptotic pathway in vitro.
Collapse
|
29
|
Scrimgeour LA, Potz BA, Aboul Gheit A, Shi G, Stanley M, Zhang Z, Sodha NR, Ahsan N, Abid MR, Sellke FW. Extracellular Vesicles Promote Arteriogenesis in Chronically Ischemic Myocardium in the Setting of Metabolic Syndrome. J Am Heart Assoc 2019; 8:e012617. [PMID: 31354010 PMCID: PMC6761642 DOI: 10.1161/jaha.119.012617] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Ischemic heart disease continues to be a leading cause of mortality in patients. Extracellular vesicles (EVs) provide a potential for treatment that may induce collateral vessel growth to increase myocardial perfusion. Methods and Results Nineteen male Yorkshire pigs were given a high‐fat diet for 4 weeks, then underwent placement of an ameroid constrictor on the left circumflex artery to induce chronic myocardial ischemia. Two weeks later, the pigs received either intramyocardial vehicle (n=6), EVs (high‐fat diet with myocardial EV injection [HVM]; n=8), or HVM and calpain inhibition (n=5). Five weeks later, myocardial function, perfusion, coronary vascular density, and cell signaling were examined. Perfusion in the collateral‐dependent myocardium was increased during rapid ventricular pacing in the HVM group in both nonischemic (P=0.04) and ischemic areas of the ventricle (P=0.05). Cardiac output and stroke volume were significantly improved in the HVM group compared with the control group during ventricular pacing (P=0.006). Increased arteriolar density was seen in the HVM group in both nonischemic and ischemic myocardium (P=0.003 for both). However, no significant changes in the capillary density were observed between the control, HVM, and HVM and calpain inhibition groups (P=0.07). The group that received EVs with oral calpain inhibition had neither increased vessel density (P>0.99) nor improvement in blood flow or cardiac function (P=0.48) when compared with the control group. Conclusions These findings suggest that EVs promote angiogenesis in areas of chronic myocardial ischemia and improve cardiac function under conditions of diet‐induced metabolic syndrome.
Collapse
Affiliation(s)
- Laura A Scrimgeour
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Brittany A Potz
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Ahmad Aboul Gheit
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Guangbin Shi
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Melissa Stanley
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Zhiqi Zhang
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Neel R Sodha
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Nagib Ahsan
- Center of Biomedical Research Excellence Center for Cancer Research Development Proteomics Core Facility Rhode Island Hospital Providence RI.,Division of Biology and Medicine Brown University Providence RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery Department of Surgery Cardiovascular Research Center Rhode Island Hospital Warren Alpert Medical School of Brown University Providence RI
| |
Collapse
|
30
|
Activation of PERK-eIF2α-ATF4 pathway contributes to diabetic hepatotoxicity: Attenuation of ER stress by Morin. Cell Signal 2019; 59:41-52. [PMID: 30877037 DOI: 10.1016/j.cellsig.2019.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 01/12/2023]
Abstract
Hyperglycemia associated ER stress has been found as a critical contributor in the pathogenesis of type 2 diabetes mellitus. However, reports regarding molecular mechanisms involved are limited. This study was aimed to identify the role of ER stress in regulating hepatic glucose metabolism and its link with oxidative stress. Further, this study explores the novel role of Morin, a flavonol, in modulating ER stress in STZ/nicotinamide induced type 2 diabetic male Wistar rats. Results demonstrate that hyperglycemia induced ER stress in rats and significantly lowered the expression of glucose transporter proteins resulting in impaired glucose metabolism during diabetes. Morin was found to downregulate PERK-eIF2α-ATF4 pathway by interacting with PERK protein as confirmed through pull-down assay. Additionally, Morin maintained the reducing environment in ER and enhanced PDI activity compared to diabetic rats. Morin prevented cell death by suppressing the expression of PERK dependent pro-apoptotic proteins including ATF4 and CHOP. Findings from this study affirm the role of ER stress in hyperglycemia induced gluco-metabolic aberrations and liver injury as confirmed by ISRIB, a standard chemical ER stress inhibitor. Notably, Morin promoted deactivation of UPR sensors and upregulated PDI activity endorsing its anti-ER stress potential which may allow the development of new therapeutic avenues to target hyperglycemic hepatotoxicity.
Collapse
|
31
|
Zheng D, Su Z, Zhang Y, Ni R, Fan GC, Robbins J, Song LS, Li J, Peng T. Calpain-2 promotes MKP-1 expression protecting cardiomyocytes in both in vitro and in vivo mouse models of doxorubicin-induced cardiotoxicity. Arch Toxicol 2019; 93:1051-1065. [DOI: 10.1007/s00204-019-02405-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
|
32
|
Chen Q, Thompson J, Hu Y, Das A, Lesnefsky EJ. Cardiac Specific Knockout of p53 Decreases ER Stress-Induced Mitochondrial Damage. Front Cardiovasc Med 2019; 6:10. [PMID: 30838215 PMCID: PMC6389610 DOI: 10.3389/fcvm.2019.00010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/30/2019] [Indexed: 11/18/2022] Open
Abstract
Endoplasmic reticulum (ER) stress contributes to cardiovascular disease including heart failure. Interactions between the ER and mitochondria during ER stress can impair the mitochondrial respiratory chain and increase cell injury. p53 is a tumor suppressor protein that regulates apoptosis. p53 contributes to the regulation of mitochondrial and ER interactions, especially during the progression of ER stress. The knockout (KO) of p53 leads to decreased injury in hearts following ischemia-reperfusion. We asked if KO of p53 can protect mitochondria during the induction of ER stress and decrease cell injury. Floxed p53 mice were crossed with mice carrying an α-myosin heavy chain cre to generate cardiac specific p53 KO mice. Thapsigargin (THAP) was used to induce ER stress in wild type (WT) and p53 KO mice. Mice were euthanized after 48 h THAP treatment. Cardiac mitochondria were isolated for functional measurement. TUNEL staining was used to assess myocyte death. In WT mice, THAP treatment decreased the rate of oxidative phosphorylation using pyruvate + malate as complex I substrates compared to vehicle-treated control. Complex I activity was also decreased in the THAP-treated WT mice. The rate of oxidative phosphorylation and complex I activity were not altered in THAP-treated p53 KO mice. The content of pyruvate dehydrogenase (PDH) α1 subunit was decreased in THAP-treated WT mice but not in p53 KO mice. ER stress led to a release of cytochrome c and apoptosis inducing factor from mitochondria into cytosol in WT but not in KO mice. Knockout of p53 also preserved mitochondrial bcl-2 content in THAP-treated mice. In WT mice, THAP treatment markedly increased cell death compared to vehicle treated hearts. In contrast, cell injury was decreased in THAP-treated p53 KO mice compared to corresponding wild type. Thus, KO of p53 decreased cell injury by protecting mitochondria during the ER stress.
Collapse
Affiliation(s)
- Qun Chen
- Division of Cardiology, Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Jeremy Thompson
- Division of Cardiology, Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Ying Hu
- Division of Cardiology, Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Anindita Das
- Division of Cardiology, Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Edward J Lesnefsky
- Division of Cardiology, Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States.,Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States.,Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States.,McGuire Department of Veterans Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
33
|
Liu ZF, Ji JJ, Zheng D, Su L, Peng T. Calpain-2 protects against heat stress-induced cardiomyocyte apoptosis and heart dysfunction by blocking p38 mitogen-activated protein kinase activation. J Cell Physiol 2018; 234:10761-10770. [PMID: 30417356 DOI: 10.1002/jcp.27750] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022]
Abstract
Cardiovascular dysfunction is a common complication among heatstroke patients, but its underlying mechanism is unclear. This study was designed to investigate the role of calpain-2 and its downstream signal pathway in heat stress-induced cardiomyocyte apoptosis and heart dysfunction. In cultured primary mouse neonatal cardiomyocytes (MNCs), heat stress (43°C for 2 hr) induced a heat-shock response, as indicated by upregulated heat-shock protein 27 (HSP27) expression and cellular apoptosis, as indicated by increased caspase-3 activity, DNA fragmentation and decreased cell viability. Meanwhile, heat stress decreased calpain activity, which was accompanied by downregulated calpain-2 expression and increased phosphorylation of p38, extraceIIuIar signaI-reguIated protein kinase (ERK1/2) and c-Jun N-terminaI kinase (JNK). Calpain-2 overexpression abrogated heat stress-induced apoptosis and phosphorylation of p38 and JNK, but not of ERK1/2. Blocking only p38 prevented heat stress-induced apoptosis in MNCs. In cardiac-specific calpain-2 overexpressing transgenic mice, p38 phosphorylation and cardiomyocyte apoptosis were decreased in the heart tissue of heatstroke mice, as revealed by western blot and terminal deoxynucleotidyl transferase dUTP nick end labelling assays, respectively. M-mode echocardiography also demonstrated that calpain-2 overexpression significantly improved heatstroke-induced decreases in ventricular end-diastolic volume and cardiac output. In conclusion, our study suggests that heat stress reduces calpain-2 expression, which then activates p38, leading to cardiomyocyte apoptosis and heart dysfunction.
Collapse
Affiliation(s)
- Zhi-Feng Liu
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China.,Department of Medicine, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Pathology, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Jing-Jing Ji
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China.,Departement of Pathophysiology, Southern Medical University, Guangzhou, China
| | - Dong Zheng
- Department of Medicine, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Pathology, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Lei Su
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China.,Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Tianqing Peng
- Department of Medicine, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Pathology, Critical Illness Research Center, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
34
|
Calpain inhibition ameliorates scald burn-induced acute lung injury in rats. BURNS & TRAUMA 2018; 6:28. [PMID: 30338266 PMCID: PMC6174571 DOI: 10.1186/s41038-018-0130-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022]
Abstract
Background The molecular pattern of severe burn-induced acute lung injury, characterized by cell structure damage and leukocyte infiltration, remains unknown. This study aimed to determine whether calpain, a protease involved in both processes, mediates severe burn-induced acute lung injury. Methods Rats received full-thickness scald burns covering 30% of the total body surface area, followed by instant fluid resuscitation. MDL28170 (Tocris Bioscience), an inhibitor of calpain, was given intravenously 1 h before or after the scald burn. The histological score, wet/dry weight ratio, and caspase-3 activity were examined to evaluate the degree of lung damage. Calpain activity and its source were detected by an assay kit and immunofluorescence staining. The proteolysis of membrane skeleton proteins α-fodrin and ankyrin-B, which are substrates of calpain, was measured by Western blot. Results Time-course studies showed that tissue damage reached a peak between 1 and 6 h post-scald burn and gradually diminished at 24 h. More importantly, calpain activity reached peak levels at 1 h and was maintained until 24 h, paralleled by lung damage to some extent. Western blot showed that the levels of the proteolyzed forms of α-fodrin and ankyrin-B correlated well with the degree of damage. MDL28170 at a dose of 3 mg/kg b. w. given 1 h before burn injury not only antagonized the increase in calpain activity but also ameliorated scald burn-induced lung injury, including the degradation of α-fodrin and ankyrin-B. Immunofluorescence images revealed calpain 1 and CD45 double-positive cells in the lung tissue of rats exposed to scald burn injury, suggesting that leukocytes were a dominant source of calpain. Furthermore, this change was blocked by MDL28170. Finally, MDL28170 given at 1 h post-scald burn injury significantly ameliorated the wet/dry weight ratio compared with burn injury alone. Conclusions Calpain, a product of infiltrating leukocytes, is a mediator of scald burn-induced acute lung injury that involves enhancement of inflammation and proteolysis of membrane skeleton proteins. Its late effects warrant further study.
Collapse
|
35
|
Li S, Ma J, Li JB, Lacefield JC, Jones DL, Peng TQ, Wei M. Over-expression of calpastatin attenuates myocardial injury following myocardial infarction by inhibiting endoplasmic reticulum stress. J Thorac Dis 2018; 10:5283-5297. [PMID: 30416776 DOI: 10.21037/jtd.2018.08.133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Ischemic heart injury activates calpains and endoplasmic reticulum (ER) stress in cardiomyocytes. This study investigated whether over-expression of calpastatin, an endogenous calpain inhibitor, protects the heart against myocardial infarction (MI) by inhibiting ER stress. Methods Mice over-expressing calpastatin (Tg-CAST) and littermate wild type (WT) mice were divided into four groups: WT-sham, Tg-CAST-sham, WT-MI, and Tg-CAST-MI, respectively. WT-sham and Tg-CAST-sham mice showed similar cardiac function at baseline. MI for 7 days impaired cardiac function in WT-MI mice, which was ameliorated in Tg-CAST-MI mice. Results Tg-CAST-MI mice exhibited significantly decreased diameter of the left ventricular cavity, scar area, and cardiac cell death compared to WT-MI mice. WT-MI mice had higher cardiac expression of C/EBP homologous protein (CHOP) and BIP, indicators of ER stress, compared to WT-sham mice, indicative of MI-induced ER stress. This increase was abolished in Tg-CAST-MI hearts. Furthermore, administration of tauroursodeoxycholic acid, an inhibitor of ER stress, reduced MI-induced expression of CHOP and BIP, scar area, and myocardial dysfunction. In an in vitro model of oxidative stress, H2O2 stimulation of H9c2 cardiomyoblasts induced calpain activation, CHOP expression, and cell death, all of which were prevented by the calpain inhibitor PD150606, as well as CHOP silencing. Conclusions Over-expression of calpastatin ameliorates MI-induced myocardial injury in mice. These protective effects of calpastatin are partially achieved through suppression of the ER stress/CHOP pathway.
Collapse
Affiliation(s)
- Shuai Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada.,Department of Medicine, Western University, London, Ontario, Canada.,Department of Pathology, Western University, London, Ontario, Canada
| | - Jian Ma
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jing-Bo Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - James C Lacefield
- Department of Electrical & Computer Engineering, Western University, London, Ontario, Canada.,Department of Medical Biophysics, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada
| | - Douglas L Jones
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada.,Department of Medicine, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada.,Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
| | - Tian-Qing Peng
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada.,Department of Medicine, Western University, London, Ontario, Canada.,Department of Pathology, Western University, London, Ontario, Canada
| | - Meng Wei
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
36
|
Lee J, Mun S, Park A, Kim D, Heun Cha B, Kang HG. Bicalutamide enhances fodrin-mediated apoptosis through calpain in LNCaP. Exp Biol Med (Maywood) 2018; 243:843-851. [PMID: 29860890 DOI: 10.1177/1535370218779780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer is the most common cancer in men, and before it progresses and metastasizes, the anticancer drug bicalutamide is often administered to patients. Many cases of androgen-dependent prostate cancer develop resistance during treatment with bicalutamide. Therefore, the effect of bicalutamide on androgen-dependent LNCaP prostate cancer cells is of clinical interest. The aim of this study was to demonstrate the effects of the anticancer drug bicalutamide on LNCaP prostate cancer cells by using a proteomics approach. Based on the results, 314 proteins were differentially expressed between the LNCaP and LNCaP treated with bicalutamide. The apoptosis pathway associated with differentially expressed proteins was shown in the Kyoto Encyclopedia of Gene and Genome pathway mapper. The Kyoto Encyclopedia of Gene and Genome pathway mapper results revealed that the fodrin-mediated apoptosis pathway is associated with the actions of bicalutamide and Western blotting was performed to validate these results. Impact statement We studied bicalutamide's anticancer action by using proteomics. The effect of bicalutamide on androgen-exposed LNCaP cells was also studied. KEGG identified >1.8-fold differentially expressed proteins between test group cells. KEGG mapper showed fodrin-mediated apoptosis involvement in bicalutamide's action. The anticancer effects of bicalutamide, which was further confirmed using Western blotting. Therefore, this drug is a potential candidate for understanding bicalutamide's effect on LNCaP and fodrin can be used as a biomarker monitoring status in metastatic carcinoma.
Collapse
Affiliation(s)
- Jiyeong Lee
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Sora Mun
- 2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| | - Arum Park
- 2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| | - Doojin Kim
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Byung Heun Cha
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Hee-Gyoo Kang
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea.,2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| |
Collapse
|
37
|
Du Y, Wang M, Liu X, Zhang J, Xu X, Xu H, Sun G, Sun X. Araloside C Prevents Hypoxia/Reoxygenation-Induced Endoplasmic Reticulum Stress via Increasing Heat Shock Protein 90 in H9c2 Cardiomyocytes. Front Pharmacol 2018; 9:180. [PMID: 29719506 PMCID: PMC5914297 DOI: 10.3389/fphar.2018.00180] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/19/2018] [Indexed: 01/06/2023] Open
Abstract
Araloside C (AsC) is a cardioprotective triterpenoid compound that is mainly isolated from Aralia elata. This study aims to determine the effects of AsC on hypoxia-reoxygenation (H/R)-induced apoptosis in H9c2 cardiomyocytes and its underlying mechanisms. Results demonstrated that pretreatment with AsC (12.5 μM) for 12 h significantly suppressed the H/R injury in H9c2 cardiomyocytes, including improving cell viability, attenuating the LDH leakage and preventing cardiomyocyte apoptosis. AsC also inhibited H/R-induced ER stress by reducing the activation of ER stress pathways (PERK/eIF2α and ATF6), and decreasing the expression of ER stress-related apoptotic proteins (CHOP and caspase-12). Moreover, AsC greatly improved the expression level of HSP90 compared with that in the H/R group. The use of HSP90 inhibitor 17-AAG and HSP90 siRNA blocked the above suppression effect of AsC on ER stress-related apoptosis caused by H/R. Taken together, AsC could reduce H/R-induced apoptosis possibly because it attenuates ER stress-dependent apoptotic pathways by increasing HSP90 expression.
Collapse
Affiliation(s)
- Yuyang Du
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| | - Xuesong Liu
- Center of Research and Development on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, China
| | - Jingyi Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| | - Xudong Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| | - Huibo Xu
- Academy of Chinese Medical Sciences of Jilin Province, Changchun, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
| |
Collapse
|
38
|
Etehadi Moghadam S, Azami Tameh A, Vahidinia Z, Atlasi MA, Hassani Bafrani H, Naderian H. Neuroprotective Effects of Oxytocin Hormone after an Experimental Stroke Model and the Possible Role of Calpain-1. J Stroke Cerebrovasc Dis 2018; 27:724-732. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.10.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 09/13/2017] [Accepted: 10/10/2017] [Indexed: 01/20/2023] Open
|
39
|
Zhang HJ, Chen RC, Sun GB, Yang LP, Zhu YD, Xu XD, Sun XB. Protective effects of total flavonoids from Clinopodium chinense (Benth.) O. Ktze on myocardial injury in vivo and in vitro via regulation of Akt/Nrf2/HO-1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 40:88-97. [PMID: 29496179 DOI: 10.1016/j.phymed.2018.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 11/30/2017] [Accepted: 01/13/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Clinopodium chinense (Benth.) O. Ktze is a traditional Chinese herbal medicine, which comprises the plant's total flavonoids. TFCC plays an important role in the treatment of cardiovascular disease. PURPOSE The aim of the study was to study the protective effects and possible mechanism of TFCC against isoproterenol (ISO)-mediated myocardial injury in vivo and anoxia/reoxygenation (A/R)-induced H9c2 cell injury in vitro. METHODS Male Sprague-Dawley (SD) rats were intragastrically pretreated with TFCC for 15 days. After 2 h of TFCC administration on days 14 and 15, a myocardial injury model was established with intragastric administration of 120 mg/kg of ISO daily for 2 days. The experiment was stopped 12 h after the last administration of the drugs. ECG recordings were taken after the treatment. Serum samples were assayed to determine the serum cardiac enzymes (e.g., creatine kinase, aspartate aminotransferase, and lactate dehydrogenase). The left ventricle was excised for histopathological examination, and myocardial homogenates were prepared to detection catalase, glutathione peroxidase, and superoxide dismutase. Reactive oxygen species (ROS), heme oxygenase-1(HO-1),and peroxidase were detected by the corresponding ELISA kits. H9c2 cells were pretreated with different concentrations of TFCC for 12 h before A/R exposure. Afterward, cell viability, LDH release, hoechst 33,342, and peromide iodine (PI) double staining, JC-1 staining, and ROS examination were determined. Western blot analyses of B-cell lymphoma-2, Bcl-2associated X protein, cleaved cysteinylaspartate specific protease-3 and-9, nuclear factor 2(Nrf2), HO-1 and serine/threonine protein kinase (AKT), and P-AKT were conducted. RESULTS The pretreatment of TFCC (10, 20, and 40 mg/kg) daily for 15 days prevented ISO-induced myocardial damage, including the decrease in serum cardiac enzymes and cardiomyocyte apoptotic index and improvement in the heart rate and vacuolation. TFCC also improved the free radical scavenging and antioxidant potential, thereby suggesting that one possible mechanism of TFCC-induced cardio protection is mediated by blocking the oxidative stress. To clarify these mechanisms, we performed the in vitro study by A/R-induced cytotoxicity model in H9c2 cells. TFCC pretreatment prevented apoptosis, increased the expression of HO-1, and enhanced the nuclear translocation of Nrf2. TFCC also activated phosphorylation of AKT, whereas the addition of LY294002, which is the pharmacologic inhibitor of PI3K, blocked the TFCC-induced Nrf2/HO-1 activation and cytoprotective effect. CONCLUSIONS TFCC protects against myocardial injury and enhances cellular antioxidant defense capacity by inducing the phosphorylation of AKT, which subsequently activated the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Hai-Jing Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Rong-Chang Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Gui-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| | - Long-Po Yang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Yin-di Zhu
- Academy of Chinese Materia Medica, Wenzhou Medical College, Wenzhou 325035, China
| | - Xu-Dong Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiao-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| |
Collapse
|
40
|
Shemarova IV, Nesterov VP, Korotkov SM, Sobol’ KV. Involvement of Ca2+ in the development of ischemic disorders of myocardial contractile function. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Wu K, Hu M, Chen Z, Xiang F, Chen G, Yan W, Peng Q, Chen X. Asiatic acid enhances survival of human AC16 cardiomyocytes under hypoxia by upregulating miR-1290. IUBMB Life 2017; 69:660-667. [PMID: 28686797 DOI: 10.1002/iub.1648] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/06/2017] [Indexed: 02/05/2023]
Abstract
Asiatic acid (AA) could attenuate ischemia/reperfusion induced myocardial apoptosis through upregulating the Akt/GSK-3β/HIF-1α pathway. HIF-3α is a negative regulator of HIF-1α, whose mRNA is a potential target of miR-1290. AA could upregulate miR-1290 in non-small-cell lung cancer A549 cells. This work aimed to investigate whether AA could inhibit hypoxia induced cardiomyocyte apoptosis through regulating the miR-1290/HIF3A/HIF-1α axis. The AC16 human myocardial cell line cultured under normoxic or hypoxic conditions was treated with various doses of AA for 24 h. Afterwards cell viability, apoptosis and the expression of miR-1290, HIF3A, and HIF1A were evaluated. Cells transfected with miR-1290 mimic or inhibitor were used to determine the role of miR-1290 in the anti-apoptosis effect of AA and the expression of HIF3A and HIF1A. Dual luciferase assay was performed to confirm miR-1290 targeting of HIF3A. HIF3A overexpression was achieved by transfection of HIF3A1 overexpressing lentivirus, and its effect on miR-1290 and AA-regulated survival of cardiomyocytes was evaluated. AA treatment protected cardiomyocytes from hypoxia-induced apoptosis and upregulated miR-1290 and HIF1A, but downregulated HIF3A under hypoxia. The protective effect of AA was abolished by miR-1290 knockdown, whereas enhanced by miR-1290 overexpression. In addition, miR-1290 knockdown increased HIF1A expression, but reduced HIF3A expression in cardiomyocytes. Dual luciferase assay confirmed miR-1290 direct targeting the 3' UTR of HIF3A. HIF3A overexpression counteracted the anti-apoptosis effect of AA or miR-1290. In conclusion, AA can protect cardiomyocytes against hypoxia-induced apoptosis through regulating the miR-1290/HIF3A/HIF-1α axis, and miR-1290 may be a potential target in the prevention of myocardial ischemia-reperfusion injury. © 2017 IUBMB Life, 69(9):660-667, 2017.
Collapse
Affiliation(s)
- Kai Wu
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Min Hu
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Zejin Chen
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Feixiang Xiang
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Guojian Chen
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Weihong Yan
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Qing Peng
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoping Chen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Zhao H, Xu M, Chu G. Association between myocardial cell apoptosis and calpain-1/caspase-3 expression in rats with hypoxic-ischemic brain damage. Mol Med Rep 2017; 15:2727-2731. [PMID: 28447745 DOI: 10.3892/mmr.2017.6341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 12/29/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the association between myocardial cell apoptosis and calpain-1/caspase-3 expression in a rat model of hypoxic-ischemic brain damage (HIBD). A total of 64 newborn rats were divided into control (n=8; sacrificed on day 7) and HIBD groups (n=56). HIBD group rats were sacrificed 2, 12 or 24 h, or 2, 3, 5 or 7 days following HIBD (n=8/group). A terminal deoxynucleotidyl transferase dUTP nick-end labeling assay was performed to detect myocardial apoptotic cells and calculate the apoptosis index (AI), reverse transcription-polymerase chain reaction was performed to detect myocardial calpain-1/caspase-3 mRNA expression levels and a western blot analysis was conducted to detect calpain‑1 protein expression levels. The correlations between calpain‑1 and caspase‑3 expression levels and AI were analyzed. The results demonstrated that apoptotic myocardial cells in the HIBD groups were markedly increased compared with the control group, with AI peaking in the day 3 group. Caspase‑3 and calpain‑1 mRNA expression levels were increased from 2 and 12 h following HIBD, respectively, with the most elevated levels in the day 2 group. Compared with the control group, calpain‑1 protein expression levels were increased from 2 h, with the greatest expression levels in the day 3 group (P<0.05). Calpain‑1 mRNA and protein (76/80 kDa) expression levels demonstrated positive linear correlations with AI (r=0.786, P=0.001; and r=0.853, P=0.001, respectively) Caspase-3 mRNA expression levels were positively correlated with AI (r=0.894; P=0.001). In conclusion, the present study demonstrated that in rats with HIBD, there is a positive correlation between increased apoptosis of myocardial cells and expression levels of calpain-1 and caspase-3.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Mei Xu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guilan Chu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
43
|
Xie Q, Su J, Jiao B, Shen L, Ma L, Qu X, Yu C, Jiang X, Xu Y, Sun L. ABT737 reverses cisplatin resistance by regulating ER-mitochondria Ca2+ signal transduction in human ovarian cancer cells. Int J Oncol 2016; 49:2507-2519. [DOI: 10.3892/ijo.2016.3733] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 10/07/2016] [Indexed: 11/05/2022] Open
|
44
|
Wu DD, Wu XH, Zhang LN. [Effect of leptin on expression of calpain-1 and Bcl-2 and apoptosis in myocardial tissue of neonatal rats after asphyxia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:1044-1049. [PMID: 27751228 PMCID: PMC7389535 DOI: 10.7499/j.issn.1008-8830.2016.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/13/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To study the effect of leptin on the expression of calcium-activated neutral protease 1 (calpain-1) and B cell lymphoma-2 (Bcl-2) and apoptosis in the myocardial tissue of neonatal rats after asphyxia. METHODS A total of 48 neonatal rats were randomly and equally divided into normal control group, asphyxia group, leptin treatment groups, and calpain-1 inhibitor (CAI-1) group. The neonatal rat model of asphyxia under normal atmospheric condition was established in all groups except the control group. For the leptin treatment groups, rats received 20, 80, and 160 μg/kg leptin by intraperitoneal injection immediately after model establishment, respectively. For the CAI-1 group, rats received 10 mg/kg CAI-1 by intraperitoneal injection immediately after model establishment. For all the groups, the myocardial tissue was collected at 2 hours after model establishment. Immunohistochemistry was used to measure the expression of calpain-1 and Bcl-2. The TUNEL method was used to evaluate apoptosis of myocardial cells. RESULTS The expression of calpain-1 and Bcl-2 and apoptosis index (AI) were significantly higher in the asphyxia group than in the normal control group (P˂0.05). The leptin treatment groups and the CAI-1 group had significantly lower expression of calpain-1, significantly lower AI, and significantly higher expression of Bcl-2 than the asphyxia group (P˂0.05). The CAI-1 group had the largest changes in all the indices compared with the asphyxia group. However, there were no significant differences in all indices between the 160 μg/kg leptin treatment group and the CAI-1 group. After asphyxia, the expression of calpain-1 was positively correlated with AI, while the expression of Bcl-2 was negatively correlated with AI and the expression of calpain-1 (P˂0.05). CONCLUSIONS Leptin reduces apoptosis of myocardial cells in asphyxiated neonatal rats by the inhibition of calpain-1 activation and upregulation of Bcl-2 expression.
Collapse
Affiliation(s)
- Dan-Dan Wu
- Department of Pediatrics, First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| | | | | |
Collapse
|
45
|
Li S, Zhang L, Ni R, Cao T, Zheng D, Xiong S, Greer PA, Fan GC, Peng T. Disruption of calpain reduces lipotoxicity-induced cardiac injury by preventing endoplasmic reticulum stress. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2023-2033. [PMID: 27523632 DOI: 10.1016/j.bbadis.2016.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/15/2016] [Accepted: 08/09/2016] [Indexed: 12/16/2022]
Abstract
Diabetes and obesity are prevalent in westernized countries. In both conditions, excessive fatty acid uptake by cardiomyocytes induces cardiac lipotoxicity, an important mechanism contributing to diabetic cardiomyopathy. This study investigated the effect of calpain disruption on cardiac lipotoxicity. Cardiac-specific capns1 knockout mice and their wild-type littermates (male, age of 4weeks) were fed a high fat diet (HFD) or normal diet for 20weeks. HFD increased body weight, altered blood lipid profiles and impaired glucose tolerance comparably in both capns1 knockout mice and their wild-type littermates. Calpain activity, cardiomyocyte cross-sectional areas, collagen deposition and triglyceride were significantly increased in HFD-fed mouse hearts, and these were accompanied by myocardial dysfunction and up-regulation of hypertrophic and fibrotic collagen genes as well as pro-inflammatory cytokines. These effects of HFD were attenuated by disruption of calpain in capns1 knockout mice. Mechanistically, deletion of capns1 in HFD-fed mouse hearts and disruption of calpain with calpain inhibitor-III, silencing of capn1, or deletion of capns1 in palmitate-stimulated cardiomyocytes prevented endoplasmic reticulum stress, apoptosis, cleavage of caspase-12 and junctophilin-2, and pro-inflammatory cytokine expression. Pharmacological inhibition of endoplasmic reticulum stress diminished palmitate-induced apoptosis and pro-inflammatory cytokine expression in cardiomyocytes. In summary, disruption of calpain prevents lipotoxicity-induced apoptosis in cardiomyocytes and cardiac injury in mice fed a HFD. The role of calpain is mediated, at least partially, through endoplasmic reticulum stress. Thus, calpain/endoplasmic reticulum stress may represent a new mechanism and potential therapeutic targets for cardiac lipotoxicity.
Collapse
Affiliation(s)
- Shengcun Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Lulu Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Rui Ni
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Critical Illness Research, Lawson Health Research Institute, Western University, London, Ontario N6A 4G5, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4G5, Canada
| | - Ting Cao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Dong Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Critical Illness Research, Lawson Health Research Institute, Western University, London, Ontario N6A 4G5, Canada; Department of Medicine, Western University, London, Ontario N6A 4G5, Canada
| | - Sidong Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Peter A Greer
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Critical Illness Research, Lawson Health Research Institute, Western University, London, Ontario N6A 4G5, Canada; Department of Medicine, Western University, London, Ontario N6A 4G5, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 4G5, Canada.
| |
Collapse
|
46
|
Zhang B, Chen Y, Shen Q, Liu G, Ye J, Sun G, Sun X. Myricitrin Attenuates High Glucose-Induced Apoptosis through Activating Akt-Nrf2 Signaling in H9c2 Cardiomyocytes. Molecules 2016; 21:molecules21070880. [PMID: 27399653 PMCID: PMC6274128 DOI: 10.3390/molecules21070880] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/26/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
Hyperglycemia, as well as diabetes mellitus, has been shown to trigger cardiac cell apoptosis. We have previously demonstrated that myricitrin prevents endothelial cell apoptosis. However, whether myricitrin can attenuate H9c2 cell apoptosis remains unknown. In this study, we established an experiment model in H9c2 cells exposed to high glucose. We tested the hypothesis that myricitrin may inhibit high glucose (HG)-induced cardiac cell apoptosis as determined by TUNEL staining. Furthermore, myricitrin promoted antioxidative enzyme production, suppressed high glucose-induced reactive oxygen species (ROS) production and decreased mitochondrial membrane potential (MMP) in H9c2 cells. This agent significantly inhibited apoptotic protein expression, activated Akt and facilitated the transcription of NF-E2-related factor 2 (Nrf2)-mediated protein (heme oxygenase-1 (HO-1) and quinone oxidoreductase 1 (NQO-1) expression as determined by Western blotting. Significantly, an Akt inhibitor (LY294002) or HO-1 inhibitor (ZnPP) not only inhibited myricitrin-induced HO-1/NQO-1 upregulation but also alleviated its anti-apoptotic effects. In summary, these observations demonstrate that myricitrin activates Nrf2-mediated anti-oxidant signaling and attenuates H9c2 cell apoptosis induced by high glucose via activation of Akt signaling.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| | - Yaping Chen
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Qiang Shen
- Center of Research and Development on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin 150076, China.
| | - Guiyan Liu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
| |
Collapse
|
47
|
Calpain inhibitor attenuates ER stress-induced apoptosis in injured spinal cord after bone mesenchymal stem cells transplantation. Neurochem Int 2016; 97:15-25. [DOI: 10.1016/j.neuint.2016.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/09/2016] [Accepted: 04/28/2016] [Indexed: 12/25/2022]
|
48
|
Folch-Puy E, Panisello A, Oliva J, Lopez A, Castro Benítez C, Adam R, Roselló-Catafau J. Relevance of Endoplasmic Reticulum Stress Cell Signaling in Liver Cold Ischemia Reperfusion Injury. Int J Mol Sci 2016; 17:807. [PMID: 27231901 PMCID: PMC4926341 DOI: 10.3390/ijms17060807] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is involved in calcium homeostasis, protein folding and lipid biosynthesis. Perturbations in its normal functions lead to a condition called endoplasmic reticulum stress (ERS). This can be triggered by many physiopathological conditions such as alcoholic steatohepatitis, insulin resistance or ischemia-reperfusion injury. The cell reacts to ERS by initiating a defensive process known as the unfolded protein response (UPR), which comprises cellular mechanisms for adaptation and the safeguarding of cell survival or, in cases of excessively severe stress, for the initiation of the cell death program. Recent experimental data suggest the involvement of ERS in ischemia/reperfusion injury (IRI) of the liver graft, which has been considered as one of major problems influencing outcome after liver transplantation. The purpose of this review is to summarize updated data on the molecular mechanisms of ERS/UPR and the consequences of this pathology, focusing specifically on solid organ preservation and liver transplantation models. We will also discuss the potential role of ERS, beyond the simple adaptive response and the regulation of cell death, in the modification of cell functional properties and phenotypic changes.
Collapse
Affiliation(s)
- Emma Folch-Puy
- Experimental Pathology Department, Instituto de Investigaciones Biomédicas de Barcelona, Spanish Research Council (IIBB-CSIC), Rosselló 161, 08036-Barcelona, Catalonia, Spain.
| | - Arnau Panisello
- Experimental Pathology Department, Instituto de Investigaciones Biomédicas de Barcelona, Spanish Research Council (IIBB-CSIC), Rosselló 161, 08036-Barcelona, Catalonia, Spain.
| | - Joan Oliva
- Department of Medicine, LaBioMed at Harbor UCLA Medical Center, Torrance, 90502 CA, USA.
| | - Alexandre Lopez
- Centre Hépatobiliaire, AP-HP Hôpital Paul Brousse, Inserm U935, Université Paris-Sud, Villejuif, 75008 Paris, France.
| | - Carlos Castro Benítez
- Centre Hépatobiliaire, AP-HP Hôpital Paul Brousse, Inserm U935, Université Paris-Sud, Villejuif, 75008 Paris, France.
| | - René Adam
- Centre Hépatobiliaire, AP-HP Hôpital Paul Brousse, Inserm U935, Université Paris-Sud, Villejuif, 75008 Paris, France.
| | - Joan Roselló-Catafau
- Experimental Pathology Department, Instituto de Investigaciones Biomédicas de Barcelona, Spanish Research Council (IIBB-CSIC), Rosselló 161, 08036-Barcelona, Catalonia, Spain.
| |
Collapse
|
49
|
Sulforaphane prevents rat cardiomyocytes from hypoxia/reoxygenation injury in vitro via activating SIRT1 and subsequently inhibiting ER stress. Acta Pharmacol Sin 2016; 37:344-53. [PMID: 26775664 DOI: 10.1038/aps.2015.130] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/26/2015] [Indexed: 02/06/2023] Open
Abstract
AIM Sulforaphane (SFN), a natural dietary isothiocyanate, is found to exert beneficial effects for cardiovascular diseases. This study aimed to investigate the mechanisms underlying the protective effects of SFN in a model of myocardial hypoxia/reoxygenation (H/R) injury in vitro. METHODS Cultured neonatal rat cardiomyocytes pretreated with SFN were subjected to 3-h hypoxia followed by 3-h reoxygenation. Cell viability and apoptosis were detected. Caspase-3 activity and mitochondrial membrane potential (ΔΨm) was measured. The expression of ER stress-related apoptotic proteins were analyzed with Western blot analyses. Silent information regulator 1 (SIRT1) activity was determined with SIRT1 deacetylase fluorometric assay kit. RESULTS SFN (0.1-5 μmol/L) dose-dependently improved the viability of cardiomyocytes, diminished apoptotic cells and suppressed caspase-3 activity. Meanwhile, SFN significantly alleviated the damage of ΔΨm and decreased the expression of ER stress-related apoptosis proteins (GRP78, CHOP and caspase-12), elevating the expression of SIRT1 and Bcl-2/Bax ratio in the cardiomyocytes. Co-treatment of the cardiomyocytes with the SIRT1-specific inhibitor Ex-527 (1 μmol/L) blocked the SFN-induced cardioprotective effects. CONCLUSION SFN prevents cardiomyocytes from H/R injury in vitro most likely via activating SIRT1 pathway and subsequently inhibiting the ER stress-dependent apoptosis.
Collapse
|
50
|
Mei M, Tang F, Lu M, He X, Wang H, Hou X, Hu J, Xu C, Han R. Astragaloside IV attenuates apoptosis of hypertrophic cardiomyocyte through inhibiting oxidative stress and calpain-1 activation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:764-773. [PMID: 26433482 DOI: 10.1016/j.etap.2015.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 09/10/2015] [Accepted: 09/13/2015] [Indexed: 06/05/2023]
Abstract
Calpain-1 activation and oxidative stress are two critical factors contributing to apoptosis of hypertrophic cardiomyocyte. Astragaloside IV (ASIV) exhibits protective effect against various heart diseases. The present study was designed to investigate whether the inhibitory effect of ASIV on isoproterenol (ISO)-induced apoptosis of hypertrophic cardiomyocyte was associated with the anti-oxidation and calpain-1 inhibition. Hypertrophy, apoptosis, mitochondrial oxidative stress and calpain-1 expression were measured in the heart tissue of Sprague-Dawley (SD) rats and H9C2 cells treated with ISO alone or combination with ASIV. The results showed that ASIV attenuated apoptotic rate, increased Bcl-2 expression, decreased Bax expression, ameliorated the integrity of mitochondrial structure and improved mitochondrial membrane potential (MMP). Moreover, ASIV combination reduced both calpain-1 protein expression and calpain activity, down-regulated mitochondrial NOX4 (mito-NOX4) expression, increased activity of mitochondrial superoxide dismutase (mito-SOD) and mitochondrial catalase (mito-CAT) compared to ISO treated alone. The results suggested that ASIV exerted anti-apoptosis effect on ISO-induced hypertrophic cardiomyocyte by attenuating oxidative stress and calpain-1 activation.
Collapse
Affiliation(s)
- Meng Mei
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| | - Futian Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| | - Xin He
- Internal Medicine-Cardiovascular Department, The First Affiliated Hospital of Liaoning Medical University, PR China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China.
| | - Xuwei Hou
- Human Anatomy Department of Liaoning Medical University, PR China
| | - Jin Hu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| | - Chonghua Xu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| | - Ronghui Han
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Drug Research Institute, Liaoning Medical University, Jinzhou, Liaoning, PR China
| |
Collapse
|