1
|
Aluja D, Delgado-Tomás S, Barrabés JA, Miró-Casas E, Ruiz-Meana M, Rodríguez-Sinovas A, Benito B, Wang J, Song LS, Ferreira-González I, Inserte J. Efficacy of a cysteine protease inhibitor compared with enalapril in murine heart failure models. iScience 2024; 27:110935. [PMID: 39381741 PMCID: PMC11458958 DOI: 10.1016/j.isci.2024.110935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Cysteine proteases calpains contribute to heart failure (HF), but it remains unknown whether their inhibition provides any benefit compared to standard pharmacological treatment for HF. Here, we characterize the pharmacological properties of NPO-2270 (NPO) as a potent inhibitor of cysteine proteases. Then, we describe that acute administration of NPO in rodent models of transient ischemia at the time of reperfusion reduces myocardial infarction, while its chronic oral administration attenuates adverse remodeling and cardiac dysfunction induced by ischemic and non-ischemic pathological stimuli more effectively than enalapril when given at the same dose. Finally, we provide evidence showing that the effects of NPO correlate with calpain inhibition and the preservation of the T-tubule morphology, due at least in part to reduced cleavage of the calpain substrate junctophilin-2. Together, our data highlight the potential of cysteine protease inhibition with NPO as a therapeutic strategy for the treatment of heart failure.
Collapse
Affiliation(s)
- David Aluja
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Sara Delgado-Tomás
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jose A. Barrabés
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Elisabet Miró-Casas
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Begoña Benito
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Jinxi Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ignacio Ferreira-González
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d’Hebron University Hospital and Research Institute, 08035 Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
2
|
Kidzeru EB, Sinkala M, Chalwa T, Matobole R, Alkelani M, Ghasemishahrestani Z, Mbandi SK, Blackburn J, Tabb DL, Adeola HA, Khumalo NP, Bayat A. Subcellular Fractionation and Metaproteogenomic Identification and Validation of Key Differentially Expressed Molecular Targets for Keloid Disease. J Invest Dermatol 2024:S0022-202X(24)01972-9. [PMID: 39122141 DOI: 10.1016/j.jid.2024.07.010] [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: 03/03/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 08/12/2024]
Abstract
Keloid disease (KD) is a common connective tissue disorder of unknown aetiopathogenesis with ill-defined treatment. Keloid scars present as exophytic fibroproliferative reticular lesions postcutaneous injury, and even though KD remains neoplastically benign, keloid lesions behave locally aggressive, invasive and expansive. To date, there is limited understanding and validation of biomarkers identified through combined proteomic and genomic evaluation of KD. Therefore, the aim in this study was to identify putative causative candidates in KD by performing a comprehensive proteomics analysis of subcellular fractions as well as the whole cell, coupled with transcriptomics data analysis of normal compared with KD fibroblasts. We then applied novel integrative bioinformatics analysis to demonstrate that NF-kB-p65 (RELA) from the cytosolic fraction and CAPN2 from the whole-cell lysate were statistically significantly upregulated in KD and associated with alterations in relevant key signaling pathways, including apoptosis. Our findings were further confirmed by showing upregulation of both RELA and CAPN2 in KD using flow cytometry and immunohistochemistry. Moreover, functional evaluation using real-time cell analysis and flow cytometry demonstrated that both omeprazole and dexamethasone inhibited the growth of KD fibroblasts by enhancing the rate of apoptosis. In conclusion, subcellular fractionation and metaproteogenomic analyses have identified, to our knowledge, 2 previously unreported biomarkers of significant relevance to keloid diagnostics and therapeutics.
Collapse
Affiliation(s)
- Elvis B Kidzeru
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa; Microbiology, Infectious Diseases, and Immunology Laboratory (LAMMII), Centre for Research on Health and Priority Pathologies (CRSPP), Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon; Division of Radiation Oncology, Department of Radiation Medicine, Groote Schuur Hospital, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Musalula Sinkala
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Temwani Chalwa
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Relebohile Matobole
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Madeha Alkelani
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Zeinab Ghasemishahrestani
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Stanley K Mbandi
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Cape Town, South Africa; Division of Immunology, Department of Pathology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Jonathan Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - David L Tabb
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa; Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Henry Ademola Adeola
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla P Khumalo
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 0] [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
|
4
|
Wang J, Chen X, Zhang L, Zheng Y, Qian J, Sun N, Ding X, Cui B. Chick early amniotic fluid component improves heart function and protects against inflammation after myocardial infarction in mice. Front Cardiovasc Med 2022; 9:1042852. [DOI: 10.3389/fcvm.2022.1042852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Myocardial infarction (MI) is the major cause of mortality around the world. We recently demonstrated that chick early amniotic fluid (ceAF) can effectively rescue ischemic heart injury, indicating that it has a therapeutic function in MI. However, its functional components and the underlying mechanisms remain to be clarified. Here, we demonstrated that a fraction of ceAF, peak 8 (P8), had a protective effect on acute MI. P8 significantly decreased cardiomyocyte cross-sectional areas and cardiomyocyte apoptosis in MI mice. Using a human embryonic stem cell-derived cardiomyocyte model, which was subjected to hypoxia and reoxygenation, mimicking MI state, we found that P8 treatment reduced apoptosis and reversed myocardial contractility. Mechanistically, P8 improved cardiac function by inhibiting NF-κB signaling and downregulating inflammatory cytokine expression. Using mass spectrometry, we identified that guanosine and deoxynucleoside were the main functional components of P8 that suppressed the inflammatory response in human embryonic stem cell-derived cardiomyocytes. Collectively, our data suggest that specific components from ceAF are promising therapeutic agents for ischemic heart injury and could be a potential supplement to current medications for MI.
Collapse
|
5
|
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: 2.5] [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
|
6
|
Aluja D, Delgado-Tomás S, Ruiz-Meana M, Barrabés JA, Inserte J. Calpains as Potential Therapeutic Targets for Myocardial Hypertrophy. Int J Mol Sci 2022; 23:ijms23084103. [PMID: 35456920 PMCID: PMC9032729 DOI: 10.3390/ijms23084103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/26/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
Despite advances in its treatment, heart failure remains a major cause of morbidity and mortality, evidencing an urgent need for novel mechanism-based targets and strategies. Myocardial hypertrophy, caused by a wide variety of chronic stress stimuli, represents an independent risk factor for the development of heart failure, and its prevention constitutes a clinical objective. Recent studies performed in preclinical animal models support the contribution of the Ca2+-dependent cysteine proteases calpains in regulating the hypertrophic process and highlight the feasibility of their long-term inhibition as a pharmacological strategy. In this review, we discuss the existing evidence implicating calpains in the development of cardiac hypertrophy, as well as the latest advances in unraveling the underlying mechanisms. Finally, we provide an updated overview of calpain inhibitors that have been explored in preclinical models of cardiac hypertrophy and the progress made in developing new compounds that may serve for testing the efficacy of calpain inhibition in the treatment of pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- David Aluja
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Sara Delgado-Tomás
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José A. Barrabés
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-934894038
| |
Collapse
|
7
|
Yin C, Ye Z, Wu J, Huang C, Pan L, Ding H, Zhong L, Guo L, Zou Y, Wang X, Wang Y, Gao P, Jin X, Yan X, Zou Y, Huang R, Gong H. Elevated Wnt2 and Wnt4 activate NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 following myocardial infarction. EBioMedicine 2021; 74:103745. [PMID: 34911029 PMCID: PMC8669316 DOI: 10.1016/j.ebiom.2021.103745] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Acute myocardial infarction (AMI)-induced excessive myocardial fibrosis exaggerates cardiac dysfunction. However, serum Wnt2 or Wnt4 level in AMI patients, and the roles in cardiac fibrosis are largely unkown. Methods AMI and non-AMI patients were enrolled to examine serum Wnt2 and Wnt4 levels by ELISA analysis. The AMI patients were followed-up for one year. MI mouse model was built by ligation of left anterior descending branch (LAD). Findings Serum Wnt2 or Wnt4 level was increased in patients with AMI, and the elevated Wnt2 and Wnt4 were correlated to adverse outcome of these patients. Knockdown of Wnt2 and Wnt4 significantly attenuated myocardial remodeling and cardiac dysfunction following experimental MI. In vitro, hypoxia enhanced the secretion and expression of Wnt2 and Wnt4 in neonatal rat cardiac myocytes (NRCMs) or fibroblasts (NRCFs). Mechanistically, the elevated Wnt2 or Wnt4 activated β-catenin /NF-κB signaling to promote pro-fibrotic effects in cultured NRCFs. In addition, Wnt2 or Wnt4 upregulated the expression of these Wnt co-receptors, frizzled (Fzd) 2, Fzd4 and (ow-density lipoprotein receptor-related protein 6 (LRP6). Further analysis revealed that Wnt2 or Wnt4 activated β-catenin /NF-κB by the co-operation of Fzd4 or Fzd2 and LRP6 signaling, respectively. Interpretation Elevated Wnt2 and Wnt4 activate β-catenin/NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 in fibroblasts, which contributes to adverse outcome of patients with AMI, suggesting that systemic inhibition of Wnt2 and Wnt4 may improve cardiac dysfunction after MI.
Collapse
Affiliation(s)
- Chao Yin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhishuai Ye
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China
| | - Jian Wu
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Chenxing Huang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Le Pan
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Huaiyu Ding
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Zhong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Guo
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yan Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiang Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Pan Gao
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xuejuan Jin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoxiang Yan
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunzeng Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China.
| | - Hui Gong
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Zhang K, Cremers MM, Wiedemann S, Poitz DM, Pfluecke C, Heinzel FR, Pieske B, Adams V, Schauer A, Winzer R, Strasser RH, Linke A, Quick S, Heidrich FM. Spatio-temporal regulation of calpain activity after experimental myocardial infarction in vivo. Biochem Biophys Rep 2021; 28:101162. [PMID: 34761128 PMCID: PMC8566776 DOI: 10.1016/j.bbrep.2021.101162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/25/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background Calpains are calcium activated cysteine proteases that play a pivotal role in the pathophysiology of cardiac remodeling. Methods Here, we performed left anterior descending coronary artery ligation in rats as a model for ischemic systolic heart failure and examined the time- and region-specific regulation of calpain-1 and calpain-2 in the left ventricular myocardium. Results Following anterior wall myocardial infarction, calpain activity was significantly increased restricted to the ischemic anterior area at days 1, 5 and 14. No changes in calpain activity at neither time point were detected in the borderzone and remote posterior area of the left ventricle. Of note, calpain activity in the infarcted anterior myocardium was regulated differentially in the acute vs. subacute and chronic phase. In the acute phase, calpain translocation to the plasma membrane and attenuation of the expression of its endogenous inhibitor, calpastatin, were identified as the driving forces. In the subacute and chronic phase, calpain activity was regulated at the level of protein expression that was shown to be essentially independent of transcriptional activity. Conclusions We conclude that myocardial infarction leads to a distinct calpain regulation pattern in the left ventricular myocardium that is region specific and time dependent. Considering the results from our previous studies, a spatio-temporal interaction between calpains and calcium dependent natriuretic peptide production in the infarcted myocardium is possible. General significance Our results shed more light in the differential regulation of calpain activity in the myocardium and might aid in the development of targeted post-infarct and/or heart failure therapeutics.
Collapse
Key Words
- AGTR1, angiotensin II receptor type 1
- Calcium
- Calpain
- Calpain-1
- Calpain-2
- Calpastatin
- Experimental myocardial infarction
- InsP3, inositol 1,4,5-trisphosphate
- InsP3R, inositol 1,4,5-trisphopshate receptor
- LAD, left anterior descending
- LVEDD, left ventricular enddiastolic diameter
- LVEF, left ventricular ejection fraction
- LVESD, left ventricular endsystolic diameter
- NF-ĸB, nuclear factor kappa B
- NT pro-ANP, N-terminal pro atrial natriuretic peptide
- SBDP, spectrin breakdown products
Collapse
Affiliation(s)
- Kun Zhang
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Melissa M Cremers
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Stephan Wiedemann
- Helios Klinikum Pirna, Department of Internal Medicine and Cardiology, Pirna, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christian Pfluecke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Volker Adams
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Antje Schauer
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Robert Winzer
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl Gustav Carus University, Technische Universität Dresden, Dresden, Germany
| | - Ruth H Strasser
- Technische Universität Dresden, Medical Faculty, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Silvio Quick
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Felix M Heidrich
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Dimethyl Fumarate Ameliorates Nucleus Pulposus Cell Dysfunction through Activating the Nrf2/HO-1 Pathway in Intervertebral Disc Degeneration. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6021763. [PMID: 34754326 PMCID: PMC8572606 DOI: 10.1155/2021/6021763] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022]
Abstract
Background Oxidative stress, inflammation, and nucleus pulposus cells (NPCs) apoptosis are involved in pathogenesis of intervertebral disc (IVD) degeneration (IVDD). Dimethyl fumarate (DMF) has been found to effectively depress oxidative stress and inflammation via the Nrf2 pathway. Hence, this project was designed to explore the underlying mechanisms of how DMF protects NPCs from damage by LPS challenge. Methods and Results CCK8 assay and flow cytometry of apoptosis indicated that DMF treatment attenuated LPS-induced NPC damage. Western blot analysis demonstrated that DMF enhanced the expressions of nuclear factor-erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in LPS-challenged NPCs. DMF treatment significantly decreased the accumulation of ROS, downregulated inflammatory cytokines (p-NF-κB, IL-1β, and TNF-α), and ER stress-associated apoptosis proteins (Bip, calpain-1, caspase-12, caspase-3, and Bax) in LPS-challenged NPCs. The level of antiapoptotic protein Bcl-2 was promoted by DMF treatment in LPS-challenged NPCs. Glutathione (GSH) assay showed that DMF treatment improved reduced to oxidized glutathione ratio in LPS-challenged NPCs. Furthermore, the results of western blot analysis indicated that in LPS-challenged NPCs, DMF treatment ameliorated the elevated levels of matrix degradation enzymes (MMP-13, aggrecanase 1) and type I collagen and the reduced levels of matrix composition (type II collagen and ACAN). However, Nrf2 knockdown abolished these protective effects of DMF. Conclusion Our data suggested that treatment with DMF mitigated LPS-induced oxidative stress, inflammation, and ER stress-associated apoptosis in NPCs via the Nrf2/HO-1 signaling pathway, thus reliving LPS-induced dysfunction of NPCs, which offered a novel potential pharmacological treatment strategy for IVDD.
Collapse
|
10
|
Dysregulated Circulating Apoptosis- and Autophagy-Related lncRNAs as Diagnostic Markers in Coronary Artery Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5517786. [PMID: 34513991 PMCID: PMC8426068 DOI: 10.1155/2021/5517786] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/03/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Objective Increasing evidence emphasizes the implications of dysregulated apoptosis and autophagy cellular processes in coronary artery disease (CAD). Herein, we aimed to explore apoptosis- and autophagy-related long noncoding RNAs (lncRNAs) in peripheral blood of CAD patients. Methods The mRNA and lncRNA expression profiles were retrieved from the Gene Expression Omnibus (GEO) database. With ∣fold change | >1.5 and adjusted p value < 0.05, differentially expressed apoptosis- and autophagy-related mRNAs were screened between CAD and healthy blood samples. Also, differentially expressed lncRNAs were identified for CAD. Using the psych package, apoptosis- and autophagy-related lncRNAs were defined with Spearson's correlation analysis. Receiver operating characteristic (ROC) curves were conducted for the assessment of the diagnosed efficacy of these apoptosis- and autophagy-related lncRNAs. Results Our results showed that 24 apoptosis- and autophagy-related mRNAs were abnormally expressed in CAD than normal controls. 12 circulating upregulated and 1 downregulated apoptosis- and autophagy-related lncRNAs were identified for CAD. The ROCs confirmed that AC004485.3 (AUC = 0.899), AC004920.3 (AUC = 0.93), AJ006998.2 (AUC = 0.776), H19 (AUC = 0.943), RP5-902P8.10 (AUC = 0.956), RP5-1114G22.2 (AUC = 0.883), RP11-247A12.1 (AUC = 0.885), RP11-288L9.4 (AUC = 0.928), RP11-344B5.2 (AUC = 0.858), RP11-452C8.1 (AUC = 0.929), RP11-565A3.1 (AUC = 0.893), and XXbac-B33L19.4 (AUC = 0.932) exhibited good performance in differentiating CAD from healthy controls. Conclusion Collectively, our findings proposed that circulating apoptosis- and autophagy-related lncRNAs could become underlying diagnostic markers for CAD in clinical practice.
Collapse
|
11
|
Calpain-Mediated Mitochondrial Damage: An Emerging Mechanism Contributing to Cardiac Disease. Cells 2021; 10:cells10082024. [PMID: 34440793 PMCID: PMC8392834 DOI: 10.3390/cells10082024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Calpains belong to the family of calcium-dependent cysteine proteases expressed ubiquitously in mammals and many other organisms. Activation of calpain is observed in diseased hearts and is implicated in cardiac cell death, hypertrophy, fibrosis, and inflammation. However, the underlying mechanisms remain incompletely understood. Recent studies have revealed that calpains target and impair mitochondria in cardiac disease. The objective of this review is to discuss the role of calpains in mediating mitochondrial damage and the underlying mechanisms, and to evaluate whether targeted inhibition of mitochondrial calpain is a potential strategy in treating cardiac disease. We expect to describe the wealth of new evidence surrounding calpain-mediated mitochondrial damage to facilitate future mechanistic studies and therapy development for cardiac disease.
Collapse
|
12
|
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: 9.3] [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
|
13
|
Guo S, Wu J, Zhou W, Liu X, Liu Y, Zhang J, Jia S, Li J, Wang H. Identification and analysis of key genes associated with acute myocardial infarction by integrated bioinformatics methods. Medicine (Baltimore) 2021; 100:e25553. [PMID: 33847684 PMCID: PMC8052032 DOI: 10.1097/md.0000000000025553] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a common disease leading threat to human health around the world. Here we aimed to explore new biomarkers and potential therapeutic targets in AMI through adopting integrated bioinformatics tools. METHODS The gene expression Omnibus (GEO) database was used to obtain genes data of AMI and no-AMI whole blood. Furthermore, differentially expressed genes (DEGs) were screened using the "Limma" package in R 3.6.1 software. Functional and pathway enrichment analyses of DEGs were performed via "Bioconductor" and "GOplot" package in R 3.6.1 software. In order to screen hub DEGs, the STRING version 11.0 database, Cytoscape and molecular complex detection (MCODE) were applied. Correlation among the hub DEGs was evaluated using Pearson's correlation analysis. RESULTS By performing DEGs analysis, 289 upregulated and 62 downregulated DEGs were successfully identified from GSE66360, respectively. And they were mainly enriched in the terms of neutrophil activation, immune response, cytokine, nuclear factor kappa-B (NF-κB) signaling pathway, IL-17 signaling pathway, and tumor necrosis factor (TNF) signaling pathway. Based on the data of protein-protein interaction (PPI), the top 10 hub genes were ranked, including interleukin-8 (CXCL8), TNF, N-formyl peptide receptor 2 (FPR2), growth-regulated alpha protein (CXCL1), transcription factor AP-1 (JUN), interleukin-1 beta (IL1B), platelet basic protein (PPBP), matrix metalloproteinase-9 (MMP9), toll-like receptor 2 (TLR2), and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G). What's more, the results of correlation analysis demonstrated that there was positive correlation between the 10 hub DEGs. CONCLUSION Ten DEGs were identified as potential candidate diagnostic biomarkers for patients with AMI in present study. However, further experiments are needed to confirm the functional pathways and hub genes associated with AMI.
Collapse
|
14
|
Ramalingam V, Hwang I. Zero valent zinc regulates adipocyte differentiation through calpain family protein and peroxisome proliferator-activated receptor gamma signaling in mouse 3T3-L1 cells. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
15
|
Liang L, Li H, Cao T, Qu L, Zhang L, Fan GC, Greer PA, Li J, Jones DL, Peng T. Calpain activation mediates microgravity-induced myocardial abnormalities in mice via p38 and ERK1/2 MAPK pathways. J Biol Chem 2020; 295:16840-16851. [PMID: 32989050 PMCID: PMC7864076 DOI: 10.1074/jbc.ra119.011890] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/22/2020] [Indexed: 12/26/2022] Open
Abstract
The human cardiovascular system has adapted to function optimally in Earth's 1G gravity, and microgravity conditions cause myocardial abnormalities, including atrophy and dysfunction. However, the underlying mechanisms linking microgravity and cardiac anomalies are incompletely understood. In this study, we investigated whether and how calpain activation promotes myocardial abnormalities under simulated microgravity conditions. Simulated microgravity was induced by tail suspension in mice with cardiomyocyte-specific deletion of Capns1, which disrupts activity and stability of calpain-1 and calpain-2, and their WT littermates. Tail suspension time-dependently reduced cardiomyocyte size, heart weight, and myocardial function in WT mice, and these changes were accompanied by calpain activation, NADPH oxidase activation, and oxidative stress in heart tissues. The effects of tail suspension were attenuated by deletion of Capns1 Notably, the protective effects of Capns1 deletion were associated with the prevention of phosphorylation of Ser-345 on p47 phox and attenuation of ERK1/2 and p38 activation in hearts of tail-suspended mice. Using a rotary cell culture system, we simulated microgravity in cultured neonatal mouse cardiomyocytes and observed decreased total protein/DNA ratio and induced calpain activation, phosphorylation of Ser-345 on p47 phox , and activation of ERK1/2 and p38, all of which were prevented by calpain inhibitor-III. Furthermore, inhibition of ERK1/2 or p38 attenuated phosphorylation of Ser-345 on p47 phox in cardiomyocytes under simulated microgravity. This study demonstrates for the first time that calpain promotes NADPH oxidase activation and myocardial abnormalities under microgravity by facilitating p47 phox phosphorylation via ERK1/2 and p38 pathways. Thus, calpain inhibition may be an effective therapeutic approach to reduce microgravity-induced myocardial abnormalities.
Collapse
Affiliation(s)
- Liwen Liang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Huili Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ting Cao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lina Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.
| | - Lulu Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Peter A Greer
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jianmin Li
- Department of Pathology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Douglas L Jones
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada; Lawson Health Research Institute of London Health Sciences Centre, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada; Lawson Health Research Institute of London Health Sciences Centre, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada.
| |
Collapse
|
16
|
Tian X, Inoue K, Zhang Y, Wang Y, Sperati CJ, Pedigo CE, Zhao T, Yan M, Groener M, Moledina DG, Ebenezer K, Li W, Zhang Z, Liebermann DA, Greene L, Greer P, Parikh CR, Ishibe S. Inhibiting calpain 1 and 2 in cyclin G associated kinase-knockout mice mitigates podocyte injury. JCI Insight 2020; 5:142740. [PMID: 33208557 PMCID: PMC7710277 DOI: 10.1172/jci.insight.142740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Evidence for reduced expression of cyclin G associated kinase (GAK) in glomeruli of patients with chronic kidney disease was observed in the Nephroseq human database, and GAK was found to be associated with the decline in kidney function. To examine the role of GAK, a protein that functions to uncoat clathrin during endocytosis, we generated podocyte-specific Gak-knockout mice (Gak-KO), which developed progressive proteinuria and kidney failure with global glomerulosclerosis. We isolated glomeruli from the mice carrying the mutation to perform messenger RNA profiling and unearthed evidence for dysregulated podocyte calpain protease activity as an important contributor to progressive podocyte damage. Treatment with calpain inhibitor III specifically inhibited calpain-1/-2 activities, mitigated the degree of proteinuria and glomerulosclerosis, and led to a striking increase in survival in the Gak-KO mice. Podocyte-specific deletion of Capns1, essential for calpain-1 and calpain-2 activities, also improved proteinuria and glomerulosclerosis in Gak-KO mice. Increased podocyte calpain activity-mediated proteolysis of IκBα resulted in increased NF-κB p65-induced expression of growth arrest and DNA-damage-inducible 45 beta in the Gak-KO mice. Our results suggest that loss of podocyte-associated Gak induces glomerular injury secondary to calcium dysregulation and aberrant calpain activation, which when inhibited, can provide a protective role.
Collapse
MESH Headings
- Animals
- Calpain/antagonists & inhibitors
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/therapy
- Female
- Glomerulosclerosis, Focal Segmental/etiology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/therapy
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Podocytes/metabolism
- Podocytes/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/physiology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/therapy
Collapse
Affiliation(s)
- Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yan Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - C. John Sperati
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher E. Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tingting Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Meihua Yan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marwin Groener
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dennis G. Moledina
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Karen Ebenezer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Wei Li
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dan A. Liebermann
- Fels Institute of Cancer Research and Molecular Biology and Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania USA
| | - Lois Greene
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Peter Greer
- Queen’s Cancer Research Institute, Kingston, Ontario, Canada
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Increased serum calpain activity is associated with HMGB1 levels in systemic sclerosis. Arthritis Res Ther 2020; 22:110. [PMID: 32393322 PMCID: PMC7216546 DOI: 10.1186/s13075-020-02195-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background Systemic sclerosis (SSc) or scleroderma is an intractable autoimmune disorder that affects multiple organs. The objectives were to investigate clinical correlations of serum calpain activity and high mobility group box 1 (HMGB1) levels with immunological and clinical traits. Methods A total of 31 patients with SSc, 20 age- and gender-matched healthy control subjects (HC), and 10 patients with other connective tissue diseases (CTD) were recruited in the study. We measured serum calpain activity and HMGB1 levels and analyzed the datasets (GSE40839, GSE48149, GSE76808, GSE81292, GSE33463, and GSE58095) from Gene Expression Omnibus (GEO) database to explore the potential mechanism by which calpain exerts its function through bioinformatics methods. Results Serum calpain activity was significantly increased in patients with SSc compared with those in HC and in patients with CTD and was correlated with serum HMGB1 levels, modified Rodnan skin score, erythrocyte sedimentation rate, mean platelet volume, and plateletcrit. Notably, serum calpain activity and HMGB1 levels in SSc patients with interstitial lung disease (ILD) were significantly higher than those in SSc patients without ILD. Serum calpain activity and HMGB1 levels could be the independent risk factors for SSc-ILD and novel biomarkers in patients with SSc. Conclusion This is the first study that reports increased serum calpain activity and the correlation between calpain and HMGB1 in patients with SSc or SSc-ILD. The serum calpain activity and HMGB1 levels may serve as measures of ILD in patients with SSc. Also, calpain and HMGB1 could be potential therapeutic targets for patients with SSc or SSc-ILD in the future.
Collapse
|
18
|
Mohsin AA, Thompson J, Hu Y, Hollander J, Lesnefsky EJ, Chen Q. Endoplasmic reticulum stress-induced complex I defect: Central role of calcium overload. Arch Biochem Biophys 2020; 683:108299. [PMID: 32061585 DOI: 10.1016/j.abb.2020.108299] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND ER (endoplasmic reticulum) stress leads to decreased complex I activity in cardiac mitochondria. The aim of the current study is to explore the potential mechanisms by which ER stress leads to the complex I defect. ER stress contributes to intracellular calcium overload and oxidative stress that are two key factors to induce mitochondrial dysfunction. Since oxidative stress is often accompanied by intracellular calcium overload during ER stress in vivo, the role of oxidative stress and calcium overload in mitochondrial dysfunction was studied using in vitro models. ER stress results in intracellular calcium overload that favors activation of calcium-dependent calpains. The contribution of mitochondrial calpain activation in ER stress-mediated complex I damage was studied. METHODS Thapsigargin (THAP) was used to induce acute ER stress in H9c2 cells and C57BL/6 mice. Exogenous calcium (25 μM) and H2O2 (100 μM) were used to induce modest calcium overload and oxidative stress in isolated mitochondria. Calpain small subunit 1 (CAPNS1) is essential to maintain calpain 1 and calpain 2 (CPN1/2) activities. Deletion of CAPNS1 eliminates the activities of CPN1/2. Wild type and cardiac-specific CAPNS1 deletion mice were used to explore the role of CPN1/2 activation in calcium-induced mitochondrial damage. RESULTS In isolated mitochondria, exogenous calcium but not H2O2 treatment led to decreased oxidative phosphorylation, supporting that calcium overload contributes a key role in the mitochondrial damage. THAP treatment of H9c2 cells decreased respiration selectively with complex I substrates. THAP treatment activated cytosolic and mitochondrial CPN1/2 in C57BL/6 mice and led to degradation of complex I subunits including NDUFS7. Calcium treatment decreased NDUFS7 content in wild type but not in CAPNS1 knockout mice. CONCLUSION ER stress-mediated activation of mitochondria-localized CPN1/2 contributes to complex I damage by cleaving component subunits.
Collapse
Affiliation(s)
- Ahmed A Mohsin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA; Radiological Techniques Department, Health and Medical Technology College-Baghdad, Middle Technical University (MTU), Iraq
| | - Jeremy Thompson
- Pauley Heart Center, Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Ying Hu
- Pauley Heart Center, Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - John Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, 25606, USA; Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, 25606, USA
| | - Edward J Lesnefsky
- Pauley Heart Center, Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA; Medical Service, McGuire Department of Veterans Affairs Medical Center, Richmond, VA, 23249, USA
| | - Qun Chen
- Pauley Heart Center, Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
19
|
Wang Y, Liu J, Kong Q, Cheng H, Tu F, Yu P, Liu Y, Zhang X, Li C, Li Y, Min X, Du S, Ding Z, Liu L. Cardiomyocyte-specific deficiency of HSPB1 worsens cardiac dysfunction by activating NFκB-mediated leucocyte recruitment after myocardial infarction. Cardiovasc Res 2020; 115:154-167. [PMID: 29982352 DOI: 10.1093/cvr/cvy163] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
Aims Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.
Collapse
Affiliation(s)
- Yana Wang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Jiali Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Cheng
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei Tu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Peng Yu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Ying Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Johnson City, TN, USA
| | - Yuehua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xinxu Min
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuya Du
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd. 300, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| |
Collapse
|
20
|
Zheng P, Chen X, Xie J, Chen X, Lin S, Ye L, Chen L, Lin J, Yu X, Zheng M. Capn4 is induced by and required for Epstein-Barr virus latent membrane protein 1 promotion of nasopharyngeal carcinoma metastasis through ERK/AP-1 signaling. Cancer Sci 2019; 111:72-83. [PMID: 31691433 PMCID: PMC6942433 DOI: 10.1111/cas.14227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/22/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Capn4, also known as CapnS1, is a member of the calpain family, which plays a crucial role in maintaining the activity and function of calpain. We previously reported that Capn4 also plays an essential role in the migration of nasopharyngeal carcinoma (NPC) cells through regulation of (MMP‐2) by nuclear factor‐kappa B activation. Epstein‐Barr virus latent membrane protein 1 (LMP1) is closely related to the malignant functions of NPC; however, the relationship between LMP1 and Capn4 in NPC remain unclear. Immunohistochemical studies showed that the level of LMP1 and Capn4 expression was high in both primary and metastatic NPC tissues, with a significantly positive correlation. We further found that LMP1 was able to upregulate the Capn4 promoter in a dose‐dependent way through the C‐terminal activation region (CTAR)1 and CTAR2 domains to activate AP‐1. Moreover, we also found that LMP1 activated AP‐1 through ERK/JNK phosphorylation. These findings indicate that Capn4 coordination with LMP1 promotes actin rearrangement and, ultimately, cellular migration. These results show that Capn4 coordination with LMP1 enhances NPC migration by increasing actin rearrangement involving ERK/JNK/AP‐1 signaling. Therapeutically, additional and more specific LMP1 and Capn4 targeted inhibitors could be exploited to treat NPC.
Collapse
Affiliation(s)
- Peichan Zheng
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xiong Chen
- Department of Medical Oncology, The 900th Hospital of the People's Liberation Army Joint Service Support Force, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianqin Xie
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xi Chen
- Department of Medical Oncology, The 900th Hospital of the People's Liberation Army Joint Service Support Force, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Shanshan Lin
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Lixiang Ye
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Lingfan Chen
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Jing Lin
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Xiangbin Yu
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Ming Zheng
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
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
|
22
|
Wang B, Zhou Q, Bi Y, Zhou W, Zeng Q, Liu Z, Liu X, Zhan Z. Phosphatase PPM1L Prevents Excessive Inflammatory Responses and Cardiac Dysfunction after Myocardial Infarction by Inhibiting IKKβ Activation. THE JOURNAL OF IMMUNOLOGY 2019; 203:1338-1347. [PMID: 31331970 DOI: 10.4049/jimmunol.1900148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/25/2019] [Indexed: 11/19/2022]
Abstract
Although the inflammatory response triggered by damage-associated molecular patterns (DAMPs) in the infarcted cardiac tissues after acute myocardial infarction (MI) contributes to cardiac repair, the unrestrained inflammation induces excessive matrix degradation and myocardial fibrosis, leading to the development of adverse remodeling and cardiac dysfunction, although the molecular mechanisms that fine tune inflammation post-MI need to be fully elucidated. Protein phosphatase Mg2+/Mn2+-dependent 1L (PPM1L) is a member of the serine/threonine phosphatase family. It is originally identified as a negative regulator of stress-activated protein kinase signaling and involved in the regulation of ceramide trafficking from the endoplasmic reticulum to Golgi apparatus. However, the role of PPM1L in MI remains unknown. In this study, we found that PPM1L transgenic mice exhibited reduced infarct size, attenuated myocardial fibrosis, and improved cardiac function. PPM1L transgenic mice showed significantly lower levels of inflammatory cytokines, including IL-1β, IL-6, TNF-α, and IL-12, in myocardial tissue. In response to DAMPs, such as HMGB1 or HSP60, released in myocardial tissue after MI, macrophages from PPM1L transgenic mice consistently produced fewer inflammatory cytokines. PPM1L-silenced macrophages showed higher levels of inflammatory cytokine production induced by DAMPs. Mechanically, PPM1L overexpression selectively inhibited the activation of NF-κB signaling in myocardial tissue post-MI and DAMP-triggered macrophages. PPM1L directly bound IKKβ and then inhibited its phosphorylation and activation, leading to impaired NF-κB signaling activation and suppressed inflammatory cytokine production. Thus, our data demonstrate that PPM1L prevents excessive inflammation and cardiac dysfunction after MI, which sheds new light on the protective regulatory mechanism underlying MI.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qingqing Zhou
- Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yong Bi
- Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200081, China
| | - Wenhui Zhou
- Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qiyan Zeng
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhongmin Liu
- Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xingguang Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China; and
| | - Zhenzhen Zhan
- Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; .,Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200081, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
23
|
Calpains mediate isoproterenol-induced hypertrophy through modulation of GRK2. Basic Res Cardiol 2019; 114:21. [DOI: 10.1007/s00395-019-0730-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/20/2019] [Indexed: 01/27/2023]
|
24
|
Cao T, Fan S, Zheng D, Wang G, Yu Y, Chen R, Song LS, Fan GC, Zhang Z, Peng T. Increased calpain-1 in mitochondria induces dilated heart failure in mice: role of mitochondrial superoxide anion. Basic Res Cardiol 2019; 114:17. [PMID: 30874894 DOI: 10.1007/s00395-019-0726-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/11/2019] [Indexed: 12/17/2022]
Abstract
We and others have reported that calpain-1 was increased in myocardial mitochondria from various animal models of heart disease. This study investigated whether constitutive up-regulation of calpain-1 restricted to mitochondria induced myocardial injury and heart failure and, if so, whether these phenotypes could be rescued by selective inhibition of mitochondrial superoxide production. Transgenic mice with human CAPN1 up-regulation restricted to mitochondria in cardiomyocytes (Tg-mtCapn1/tTA) were generated and characterized with low and high over-expression of transgenic human CAPN1 restricted to mitochondria, respectively. Transgenic up-regulation of mitochondria-targeted CAPN1 dose-dependently induced cardiac cell death, adverse myocardial remodeling, heart failure, and early death in mice, the changes of which were associated with mitochondrial dysfunction and mitochondrial superoxide generation. Importantly, a daily injection of mitochondria-targeted superoxide dismutase mimetics mito-TEMPO for 1 month starting from age 2 months attenuated cardiac cell death, adverse myocardial remodeling and heart failure, and reduced mortality in Tg-mtCapn1/tTA mice. In contrast, administration of TEMPO did not achieve similar cardiac protection in transgenic mice. Furthermore, transgenic up-regulation of mitochondria-targeted CAPN1 induced a reduction of ATP5A1 protein and ATP synthase activity in hearts. In cultured cardiomyocytes, increased calpain-1 in mitochondria promoted mitochondrial permeability transition pore (mPTP) opening and induced cell death, which were prevented by over-expression of ATP5A1, mito-TEMPO or cyclosporin A, an inhibitor of mPTP opening. In conclusion, this study has provided direct evidence demonstrating that increased mitochondrial calpain-1 is an important mechanism contributing to myocardial injury and heart failure by disrupting ATP synthase, and promoting mitochondrial superoxide generation and mPTP opening.
Collapse
Affiliation(s)
- Ting Cao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Shuai Fan
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Dong Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4S2, Canada
- Department of Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada
| | - Grace Wang
- Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yong Yu
- Shanghai Institute of Cardiovascular Diseases, Shanghai Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ruizhen Chen
- Shanghai Institute of Cardiovascular Diseases, Shanghai Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zhuxu Zhang
- Department of Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4S2, Canada.
- Department of Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada.
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, N6A 4S2, Canada.
| |
Collapse
|
25
|
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: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
|
26
|
Han Q, Liu Q, Zhang H, Lu M, Wang H, Tang F, Zhang Y. Simvastatin Improves Cardiac Hypertrophy in Diabetic Rats by Attenuation of Oxidative Stress and Inflammation Induced by Calpain-1-Mediated Activation of Nuclear Factor-κB (NF-κB). Med Sci Monit 2019; 25:1232-1241. [PMID: 30767945 PMCID: PMC6383435 DOI: 10.12659/msm.913244] [Citation(s) in RCA: 13] [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: 09/17/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Simvastatin, an HMG-CoA reductase inhibitor, has been reported to exert multiple protective effects on the cardiovascular system. However, the molecular mechanism remains to be examined. The present study was designed to study the effects of simvastatin on cardiac hypertrophy in diabetic rats and to explore its potential mechanism. MATERIAL AND METHODS Sprague-Dawley rats were assigned into a control (Con) group, a streptozotocin (STZ) group, and a STZ+simvastatin (STZ+SIM) group. The level of reactive oxygen species (ROS) was measured by using dihydroethidium (DHE) staining. The protein expressions of p65, IκBα, vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), calpain-1, and endothelial nitric oxide synthase (eNOS) were examined by Western blot analysis. qPCR was used to detect the levels of brain natriuretic peptide (BNP) and atrial natriuretic peptide (ANP). RESULTS Simvastatin improved the cardiac hypertrophy of diabetic rats, as demonstrated by decreases in the ratios of left ventricular weight/body weight (LVW/BW) and heart weight/body weight (HW/BW) and by the downregulation of mRNA expression of BNP and ANP in the heart tissue. Simvastatin decreased the protein expressions of VCAM-1, ICAM-1, IL-6, and TNF-α, increased eNOS protein expression, and limited an increase in ROS levels in the heart tissue. Simvastatin increased IkBa protein expression in cytoplasm and inhibited the translocation of p65, the subunit of nuclear factor-κB (NF-κB) to the nucleus from the cytoplasm of the heart tissue. Furthermore, simvastatin attenuated the activity of calpain and calpain-1 protein expression in heart tissue. CONCLUSIONS Simvastatin attenuates cardiac hypertrophy in diabetic rats, which might be due to the attenuation of oxidative stress and inflammation induced by calpain-1-mediated activation of NF-κB.
Collapse
Affiliation(s)
- Qianqian Han
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Qianqian Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Hui Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Futian Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Yingjie Zhang
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| |
Collapse
|
27
|
Capn4 expression is modulated by microRNA-520b and exerts an oncogenic role in prostate cancer cells by promoting Wnt/β-catenin signaling. Biomed Pharmacother 2018; 108:467-475. [DOI: 10.1016/j.biopha.2018.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/28/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
|
28
|
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: 3.3] [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
|
29
|
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: 0.5] [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
|
30
|
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: 1.5] [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
|
31
|
Wang Y, Chen B, Huang CK, Guo A, Wu J, Zhang X, Chen R, Chen C, Kutschke W, Weiss RM, Boudreau RL, Margulies KB, Hong J, Song LS. Targeting Calpain for Heart Failure Therapy: Implications From Multiple Murine Models. JACC Basic Transl Sci 2018; 3:503-517. [PMID: 30175274 PMCID: PMC6115647 DOI: 10.1016/j.jacbts.2018.05.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/20/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022]
Abstract
Calpain is hyperactivated in human failing hearts and rodent heart failure models of different etiologies. Inhibition of calpain activity with MDL-28170 protects against cardiac dysfunction by preserving JP2 expression and T-tubule ultrastructural integrity in murine models of heart failure. Overexpression of JP2 delays the onset of early cardiac sudden death and heart failure, induced by calpain overactivation.
Heart failure remains a major cause of morbidity and mortality in developed countries. There is still a strong need to devise new mechanism-based treatments for heart failure. Numerous studies have suggested the importance of the Ca2+-dependent protease calpain in cardiac physiology and pathology. However, no drugs are currently under development or testing in human patients to target calpain for heart failure treatment. Herein the data demonstrate that inhibition of calpain activity protects against deleterious ultrastructural remodeling and cardiac dysfunction in multiple rodent models of heart failure, providing compelling evidence that calpain inhibition is a promising therapeutic strategy for heart failure treatment.
Collapse
Key Words
- CAPN1-OE, calpain-1 overexpressing
- E-C coupling, excitation-contraction coupling
- EF, ejection fraction
- IP, intraperitoneally
- ISO, isoproterenol
- JP2, junctophilin-2
- JP2-OE, junctophilin-2 overexpressing
- LV, left ventricle/ventricular
- MI, myocardial infarction
- RV, right ventricular
- SR, sarcoplasmic reticulum
- T-tubule, transverse tubule
- T-tubules
- TAB, transverse aortic banding
- TTpower, strength of regularity of the T-tubule system
- WT, wild-type
- calcium
- calpain
- excitation-contraction coupling
- heart failure
Collapse
Affiliation(s)
- Yihui Wang
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China.,Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa.,Department of Veterans Affairs Medical Center, Iowa City, Iowa
| | - Chun-Kai Huang
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China.,Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Jennifer Wu
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Xiaoming Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Rong Chen
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China.,Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Cheng Chen
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China.,Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - William Kutschke
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Robert M Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Ryan L Boudreau
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa
| | - Kenneth B Margulies
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jiang Hong
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine & François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine; Iowa City, Iowa.,Department of Veterans Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
32
|
Poncelas M, Inserte J, Aluja D, Hernando V, Vilardosa U, Garcia-Dorado D. Delayed, oral pharmacological inhibition of calpains attenuates adverse post-infarction remodelling. Cardiovasc Res 2018; 113:950-961. [PMID: 28460013 DOI: 10.1093/cvr/cvx073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/27/2017] [Indexed: 01/12/2023] Open
Abstract
Calpains activate during myocardial ischemia-reperfusion and contribute to reperfusion injury. Studies in transgenic animals with altered calpain/calpastatin system subjected to permanent ischemia suggest that calpains are also involved in post-infarction remodelling and heart failure. Aims To determine whether delayed oral administration of the calpain inhibitor SNJ-1945 reduces adverse myocardial remodelling and dysfunction following transient coronary occlusion. Methods and results Male Sprague-Dawley rats were subjected to 30 min of ischemia followed by 21 days of reperfusion and received the calpain inhibitor SNJ-1945 intraperitoneally at the onset of reperfusion (Acute group), orally starting after 24 h of reperfusion and for 14 days (Chronic group), or the combination of both treatments. Calpain-1 and calpain-2 protein content increased and correlated with higher calpain activity in control hearts. Administration of SNJ-1945 attenuated calpain activation, and reduced scar expansion, ventricular dilation and dysfunction in both acute and chronic groups. Acute treatment reduced infarct size in hearts reperfused for 24 h and inflammation measured after 3 days. Delayed, chronic oral administration of SNJ-1945 attenuated inflammation, cardiomyocyte hypertrophy and collagen infiltration in the non-infarcted myocardium at 21 days in correlation with increased levels of IĸB and reduced NF-ĸB activation. In cultured fibroblasts, SNJ-1945 attenuated TGF-β1-induced fibroblast activation. Conclusions Our data demonstrate for the first time that long-term calpain inhibition is possible with delayed oral treatment, attenuates adverse post-infarction remodelling, likely through prevention of NF-ĸB activation, and may be a promising therapeutic intervention to prevent adverse remodelling and heart failure in patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Marcos Poncelas
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
| | - Javier Inserte
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
- CIBERCV, Spain
| | - David Aluja
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
| | - Victor Hernando
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
| | - Ursula Vilardosa
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
| | - David Garcia-Dorado
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron University Hospital and Research Institute VHIR, Universitat Autónoma de Barcelona, Passeig Vall d'Hebron 119-129 08035 Barcelona, Spain
- CIBERCV, Spain
| |
Collapse
|
33
|
Roumans NJT, Wang P, Vink RG, van Baak MA, Mariman ECM. Combined Analysis of Stress- and ECM-Related Genes in Their Effect on Weight Regain. Obesity (Silver Spring) 2018; 26:492-498. [PMID: 29399976 DOI: 10.1002/oby.22093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/14/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE During weight loss, the volume of adipocytes decreases, leading to stress because of the misfit between the cell contents and the surrounding extracellular matrix (ECM). This stress can be resolved by remodeling the ECM or the restorage of triglycerides within the adipocytes. The objective of this study was to investigate the existence of a connection between stress-related and ECM-related genes that is associated with weight regain. METHODS Thirty-one participants with overweight or obesity followed a 5-week very-low-calorie diet (500 kcal/d) with a subsequent 4-week weight-stable diet (WS), and then an uncontrolled 9-month follow-up. Adipose tissue biopsies were collected for microarray analysis. A correlation and interaction analysis was performed with the weight regain percentage (WR%) ([weight after follow-up - weight after WS] ÷ weight after WS × 100%) by using two gene sets that were previously defined as "stress-related" (n = 107) and "ECM-related" genes (n = 277). RESULTS During WS, a coexpression network of 8 stress-related genes and 15 ECM-related genes correlating with WR% could be constructed, with links to multiple biological processes. Interaction analysis between stress- and ECM-related genes revealed that several gene combinations were highly related to weight regain. CONCLUSIONS Our findings underscore the importance of the connection between stress- and ECM-related genes in the risk for weight regain.
Collapse
Affiliation(s)
- Nadia J T Roumans
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ping Wang
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Roel G Vink
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marleen A van Baak
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
34
|
Minocycline improves cardiac function after myocardial infarction in rats by inhibiting activation of PARP-1. Biomed Pharmacother 2018; 97:1119-1124. [DOI: 10.1016/j.biopha.2017.10.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 11/21/2022] Open
|
35
|
Muniappan L, Javidan A, Jiang W, Mohammadmoradi S, Moorleghen JJ, Katz WS, Balakrishnan A, Howatt DA, Subramanian V. Calpain Inhibition Attenuates Adipose Tissue Inflammation and Fibrosis in Diet-induced Obese Mice. Sci Rep 2017; 7:14398. [PMID: 29089532 PMCID: PMC5663911 DOI: 10.1038/s41598-017-14719-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/17/2017] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue macrophages have been proposed as a link between obesity and insulin resistance. However, the mechanisms underlying these processes are not completely defined. Calpains are calcium-dependent neutral cysteine proteases that modulate cellular function and have been implicated in various inflammatory diseases. To define whether activated calpains influence diet-induced obesity and adipose tissue macrophage accumulation, mice that were either wild type (WT) or overexpressing calpastatin (CAST Tg), the endogenous inhibitor of calpains were fed with high (60% kcal) fat diet for 16 weeks. CAST overexpression did not influence high fat diet-induced body weight and fat mass gain throughout the study. Calpain inhibition showed a transient improvement in glucose tolerance at 5 weeks of HFD whereas it lost this effect on glucose and insulin tolerance at 16 weeks HFD in obese mice. However, CAST overexpression significantly reduced adipocyte apoptosis, adipose tissue collagen and macrophage accumulation as detected by TUNEL, Picro Sirius and F4/80 immunostaining, respectively. CAST overexpression significantly attenuated obesity-induced inflammatory responses in adipose tissue. Furthermore, calpain inhibition suppressed macrophage migration to adipose tissue in vitro. The present study demonstrates a pivotal role for calpains in mediating HFD-induced adipose tissue remodeling by influencing multiple functions including apoptosis, fibrosis and inflammation.
Collapse
Affiliation(s)
- Latha Muniappan
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Aida Javidan
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Weihua Jiang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | | | | | - Wendy S Katz
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Anju Balakrishnan
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Deborah A Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Venkateswaran Subramanian
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.
- Department of Physiology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
36
|
Abstract
In most patients with chronic heart failure (HF), levels of circulating cytokines are elevated and the elevated cytokine levels correlate with the severity of HF and prognosis. Various stresses induce subcellular component abnormalities, such as mitochondrial damage. Damaged mitochondria induce accumulation of reactive oxygen species and apoptogenic proteins, and subcellular inflammation. The vicious cycle of subcellular component abnormalities, inflammatory cell infiltration and neurohumoral activation induces cardiomyocyte injury and death, and cardiac fibrosis, resulting in cardiac dysfunction and HF. Quality control mechanisms at both the protein and organelle levels, such as elimination of apoptogenic proteins and damaged mitochondria, maintain cellular homeostasis. An imbalance between protein synthesis and degradation is likely to result in cellular dysfunction and disease. Three major protein degradation systems have been identified, namely the cysteine protease system, autophagy, and the ubiquitin proteasome system. Autophagy was initially believed to be a non-selective process. However, recent studies have described the process of selective mitochondrial autophagy, known as mitophagy. Elimination of damaged mitochondria by autophagy is important for maintenance of cellular homeostasis. DNA and RNA degradation systems also play a critical role in regulating inflammation and maintaining cellular homeostasis mediated by damaged DNA clearance and post-transcriptional regulation, respectively. This review discusses some recent advances in understanding the role of sterile inflammation and degradation systems in HF.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| |
Collapse
|
37
|
Ji J, Su L, Liu Z. Critical role of calpain in inflammation. Biomed Rep 2016; 5:647-652. [PMID: 28101338 DOI: 10.3892/br.2016.785] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 09/14/2016] [Indexed: 02/07/2023] Open
Abstract
Calpains are a family of cysteine proteases, implicated in a wide range of cellular calcium-regulated functions. Evidence from previous studies using an inhibitor of calpain indicates that calpain activation is involved in the process of numerous inflammation-associated diseases. As a result of in-depth studies, calpains have been proposed to influence the process of inflammation via a variety of mechanisms. The aim of the present study is to provide an overview of recent reports regarding the role of calpain in the process of inflammation, including regulation of immune cell migration, modulation of the activation of inflammatory mediators, degradation of certain associated proteins and induction of cell apoptosis. Understanding these mechanisms may contribute to the investigation of novel therapeutic targets for inflammation-associated diseases.
Collapse
Affiliation(s)
- Jingjing Ji
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China; Graduate School, Guangzhou Medical University, Guangzhou, Guangdong 510010, P.R. China
| | - Lei Su
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China; Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Zhifeng Liu
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China; Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
38
|
Li M, Su Y, Yu Y, Yu Y, Wang X, Zou Y, Ge J, Chen R. Dual roles of calpain in facilitating Coxsackievirus B3 replication and prompting inflammation in acute myocarditis. Int J Cardiol 2016; 221:1123-31. [PMID: 27472894 PMCID: PMC7114300 DOI: 10.1016/j.ijcard.2016.07.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/17/2016] [Accepted: 07/08/2016] [Indexed: 01/14/2023]
Abstract
Background Viral myocarditis (VMC) treatment has long been lacking of effective methods. Our former studies indicated roles of calpain in VMC pathogenesis. This study aimed at verifying the potential of calpain in Coxsackievirus B3 (CVB3)-induced myocarditis treatment. Methods A transgenic mouse overexpressing the endogenous calpain inhibitor, calpastatin, was introduced in the study. VMC mouse model was established via intraperitoneal injection of CVB3 in transgenic and wild mouse respectively. Myocardial injury was assayed histologically (HE staining and pathology grading) and serologically (myocardial damage markers of CK-MB and cTnI). CVB3 replication was observed in vivo and in vitro via the capsid protein VP1 detection or virus titration. Inflammation/fibrotic factors of MPO, perforin, IFNγ, IL17, Smad3 and MMP2 were evaluated using western blot or immunohistology stain. Role of calpain in regulating fibroblast migration was studied in scratch assays. Results Calpastatin overexpression ameliorated myocardial injury induced by CVB3 infection significantly in transgenic mouse indicated by reduced peripheral CK-MB and cTnI levels and improved histology injury. Comparing with CVB3-infected wild type mouse, the transgenic mouse heart tissue carried lower virus load. The inflammation factors of MPO, perforin, IFNγ and IL17 were down-regulated accompanied with fibrotic agents of Smad3 and MMP2 inhibition. And calpain participated in the migration of fibroblasts in vitro, which further proves its role in regulating fibrosis. Conclusion Calpain plays dual roles of facilitating CVB3 replication and inflammation promotion. Calpain inhibition in CVB3-induced myocarditis showed significant treatment effect. Calpain might be a novel target for VMC treatment in clinical practices. Calpain is involved in virus replication in myocarditis. Calpain mediates inflammation infiltration in myocarditis. Calpain might be a candidate for viral myocarditis treatment.
Collapse
Affiliation(s)
- Minghui Li
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yangang Su
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Yu
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Yu
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinggang Wang
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yunzeng Zou
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Ruizhen Chen
- Department of Cardiovascular Diseases, Key Laboratory of Viral Heart Diseases, Ministry of Public Health, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
39
|
Mitochondrial function in hypoxic ischemic injury and influence of aging. Prog Neurobiol 2016; 157:92-116. [PMID: 27321753 DOI: 10.1016/j.pneurobio.2016.06.006] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 03/30/2016] [Accepted: 06/12/2016] [Indexed: 12/11/2022]
Abstract
Mitochondria are a major target in hypoxic/ischemic injury. Mitochondrial impairment increases with age leading to dysregulation of molecular pathways linked to mitochondria. The perturbation of mitochondrial homeostasis and cellular energetics worsens outcome following hypoxic-ischemic insults in elderly individuals. In response to acute injury conditions, cellular machinery relies on rapid adaptations by modulating posttranslational modifications. Therefore, post-translational regulation of molecular mediators such as hypoxia-inducible factor 1α (HIF-1α), peroxisome proliferator-activated receptor γ coactivator α (PGC-1α), c-MYC, SIRT1 and AMPK play a critical role in the control of the glycolytic-mitochondrial energy axis in response to hypoxic-ischemic conditions. The deficiency of oxygen and nutrients leads to decreased energetic reliance on mitochondria, promoting glycolysis. The combination of pseudohypoxia, declining autophagy, and dysregulation of stress responses with aging adds to impaired host response to hypoxic-ischemic injury. Furthermore, intermitochondrial signal propagation and tissue wide oscillations in mitochondrial metabolism in response to oxidative stress are emerging as vital to cellular energetics. Recently reported intercellular transport of mitochondria through tunneling nanotubes also play a role in the response to and treatments for ischemic injury. In this review we attempt to provide an overview of some of the molecular mechanisms and potential therapies involved in the alteration of cellular energetics with aging and injury with a neurobiological perspective.
Collapse
|
40
|
Howatt DA, Balakrishnan A, Moorleghen JJ, Muniappan L, Rateri DL, Uchida HA, Takano J, Saido TC, Chishti AH, Baud L, Subramanian V. Leukocyte Calpain Deficiency Reduces Angiotensin II-Induced Inflammation and Atherosclerosis But Not Abdominal Aortic Aneurysms in Mice. Arterioscler Thromb Vasc Biol 2016; 36:835-45. [PMID: 26966280 DOI: 10.1161/atvbaha.116.307285] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 02/27/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Angiotensin II (AngII) infusion profoundly increases activity of calpains, calcium-dependent neutral cysteine proteases, in mice. Pharmacological inhibition of calpains attenuates AngII-induced aortic medial macrophage accumulation, atherosclerosis, and abdominal aortic aneurysm in mice. However, the precise functional contribution of leukocyte-derived calpains in AngII-induced vascular pathologies has not been determined. The purpose of this study was to determine whether calpains expressed in bone marrow (BM)-derived cells contribute to AngII-induced atherosclerosis and aortic aneurysms in hypercholesterolemic mice. APPROACH AND RESULTS To study whether leukocyte calpains contributed to AngII-induced aortic pathologies, irradiated male low-density lipoprotein receptor(-/-) mice were repopulated with BM-derived cells that were either wild-type or overexpressed calpastatin, the endogenous inhibitor of calpains. Mice were fed a fat-enriched diet and infused with AngII (1000 ng/kg per minute) for 4 weeks. Overexpression of calpastatin in BM-derived cells significantly attenuated AngII-induced atherosclerotic lesion formation in aortic arches, but had no effect on aneurysm formation. Using either BM-derived cells from calpain-1-deficient mice or mice with leukocyte-specific calpain-2 deficiency generated using cre-loxP recombination technology, further studies demonstrated that independent deficiency of either calpain-1 or -2 in leukocytes modestly attenuated AngII-induced atherosclerosis. Calpastatin overexpression significantly attenuated AngII-induced inflammatory responses in macrophages and spleen. Furthermore, calpain inhibition suppressed migration and adhesion of macrophages to endothelial cells in vitro. Calpain inhibition also significantly decreased hypercholesterolemia-induced atherosclerosis in the absence of AngII. CONCLUSIONS The present study demonstrates a pivotal role for BM-derived calpains in mediating AngII-induced atherosclerosis by influencing macrophage function.
Collapse
Affiliation(s)
- Deborah A Howatt
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Anju Balakrishnan
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Jessica J Moorleghen
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Latha Muniappan
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Debra L Rateri
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Haruhito A Uchida
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Jiro Takano
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Takaomi C Saido
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Athar H Chishti
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Laurent Baud
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.)
| | - Venkateswaran Subramanian
- From the Saha Cardiovascular Research Center (D.A.H., A.B., J.J.M., L.M., D.L.R.), and Department of Physiology (V.S.), University of Kentucky, Lexington; Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan (H.A.U.); Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan (J.T., T.C.S.); Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA (A.H.C.); and INSERM, Université Pierre et Marie Curie-Paris, Paris, France (L.B.).
| |
Collapse
|
41
|
Ding YF, Peng YR, Shen H, Shu L, Wei YJ. Gualou Xiebai decoction inhibits cardiac dysfunction and inflammation in cardiac fibrosis rats. Altern Ther Health Med 2016; 16:49. [PMID: 26846090 PMCID: PMC4743121 DOI: 10.1186/s12906-016-1012-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023]
Abstract
Background Gualou Xiebai Decoction (GXD) is a well-known traditional Chinese recipe. It has been used to treat cardiovascular disorders for nearly two thousand years. But there is a lack of reports on cardiac fibrosis and underlying mechanism. Methods Myocardial infarction was performed by ligation of left anterior descending coronary artery (LAD) in male Wistar rats. Rats with myocardial infarction were treated with GXD (1.14 g/kg, 4.53 g/kg) daily for 4 weeks. Cardiac function was evaluated by echocardiography. Hemodynamic parameters and infarct size were measured in each group. Myocardial enzymes were examined by biochemical tests. Inflammatory cytokines were assessed by ELISA, and interrelated proteins were detected by western blot. Results Cardiac function was significantly improved in GXD-treatment rats after myocardial infarction (MI), which was accompanied with decreased infarct size. Administration of GXD to myocardial fibrosis rats significantly ameliorated the activities of AST, LDH and CK-MB in serum. The increase in inflammatory factors (TNF-α, IL-1β) were markedly reduced upon GXD treatment. Furthermore, the inflammatory mediators (NF-κB p65, TNF-α, MCP-1) were down-regulated by GXD in the myocardial fibrosis rats. Conclusions Treatment with GXD improved cardiac function induced by myocardial fibrosis by inhibiting expression of inflammatory mediators associated with NF-κB.
Collapse
|
42
|
Ni R, Zheng D, Xiong S, Hill DJ, Sun T, Gardiner RB, Fan GC, Lu Y, Abel ED, Greer PA, Peng T. Mitochondrial Calpain-1 Disrupts ATP Synthase and Induces Superoxide Generation in Type 1 Diabetic Hearts: A Novel Mechanism Contributing to Diabetic Cardiomyopathy. Diabetes 2016; 65:255-68. [PMID: 26470784 PMCID: PMC4686953 DOI: 10.2337/db15-0963] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/07/2015] [Indexed: 02/05/2023]
Abstract
Calpain plays a critical role in cardiomyopathic changes in type 1 diabetes (T1D). This study investigated how calpain regulates mitochondrial reactive oxygen species (ROS) generation in the development of diabetic cardiomyopathy. T1D was induced in transgenic mice overexpressing calpastatin, in mice with cardiomyocyte-specific capn4 deletion, or in their wild-type littermates by injection of streptozotocin. Calpain-1 protein and activity in mitochondria were elevated in diabetic mouse hearts. The increased mitochondrial calpain-1 was associated with an increase in mitochondrial ROS generation and oxidative damage and a reduction in ATP synthase-α (ATP5A1) protein and ATP synthase activity. Genetic inhibition of calpain or upregulation of ATP5A1 increased ATP5A1 and ATP synthase activity, prevented mitochondrial ROS generation and oxidative damage, and reduced cardiomyopathic changes in diabetic mice. High glucose concentration induced ATP synthase disruption, mitochondrial superoxide generation, and cell death in cardiomyocytes, all of which were prevented by overexpression of mitochondria-targeted calpastatin or ATP5A1. Moreover, upregulation of calpain-1 specifically in mitochondria induced the cleavage of ATP5A1, superoxide generation, and apoptosis in cardiomyocytes. In summary, calpain-1 accumulation in mitochondria disrupts ATP synthase and induces ROS generation, which promotes diabetic cardiomyopathy. These findings suggest a novel mechanism for and may have significant implications in diabetic cardiac complications.
Collapse
Affiliation(s)
- Rui Ni
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China Department of Medicine, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada Department of Pathology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Dong Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China Department of Medicine, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada Department of Pathology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Sidong Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - David J Hill
- Department of Medicine, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Tao Sun
- Department of Medicine, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Richard B Gardiner
- Department of Biology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Peter A Greer
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China Department of Medicine, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada Department of Pathology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
43
|
Wan F, Letavernier E, Le Saux CJ, Houssaini A, Abid S, Czibik G, Sawaki D, Marcos E, Dubois-Rande JL, Baud L, Adnot S, Derumeaux G, Gellen B. Calpastatin overexpression impairs postinfarct scar healing in mice by compromising reparative immune cell recruitment and activation. Am J Physiol Heart Circ Physiol 2015; 309:H1883-93. [PMID: 26453333 DOI: 10.1152/ajpheart.00594.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/03/2015] [Indexed: 12/15/2022]
Abstract
The activation of the calpain system is involved in the repair process following myocardial infarction (MI). However, the impact of the inhibition of calpain by calpastatin, its natural inhibitor, on scar healing and left ventricular (LV) remodeling is elusive. Male mice ubiquitously overexpressing calpastatin (TG) and wild-type (WT) controls were subjected to an anterior coronary artery ligation. Mortality at 6 wk was higher in TG mice (24% in WT vs. 44% in TG, P < 0.05) driven by a significantly higher incidence of cardiac rupture during the first week post-MI, despite comparable infarct size and LV dysfunction and dilatation. Calpain activation post-MI was blunted in TG myocardium. In TG mice, inflammatory cell infiltration and activation were reduced in the infarct zone (IZ), particularly affecting M2 macrophages and CD4(+) T cells, which are crucial for scar healing. To elucidate the role of calpastatin overexpression in macrophages, we stimulated peritoneal macrophages obtained from TG and WT mice in vitro with IL-4, yielding an abrogated M2 polarization in TG but not in WT cells. Lymphopenic Rag1(-/-) mice receiving TG splenocytes before MI demonstrated decreased T-cell recruitment and M2 macrophage activation in the IZ day 5 after MI compared with those receiving WT splenocytes. Calpastatin overexpression prevented the activation of the calpain system after MI. It also impaired scar healing, promoted LV rupture, and increased mortality. Defective scar formation was associated with blunted CD4(+) T-cell and M2-macrophage recruitment.
Collapse
Affiliation(s)
- Feng Wan
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Emmanuel Letavernier
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Claude Jourdan Le Saux
- Department of Medicine/Cardiology Division, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Amal Houssaini
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Shariq Abid
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Gabor Czibik
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Daigo Sawaki
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Elisabeth Marcos
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Jean-Luc Dubois-Rande
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Laurent Baud
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Serge Adnot
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Geneviève Derumeaux
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Barnabas Gellen
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France; Department of Cardiology, Poitiers University Hospital, F-86000, Poitiers, France
| |
Collapse
|
44
|
Chen Q, Lesnefsky EJ. Heart mitochondria and calpain 1: Location, function, and targets. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2372-8. [PMID: 26259540 DOI: 10.1016/j.bbadis.2015.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/17/2015] [Accepted: 08/06/2015] [Indexed: 12/22/2022]
Abstract
Calpain 1 is an ubiquitous Ca(2+)-dependent cysteine protease. Although calpain 1 has been found in cardiac mitochondria, the exact location within mitochondrial compartments and its function remain unclear. The aim of the current review is to discuss the localization of calpain 1 in different mitochondrial compartments in relationship to its function, especially in pathophysiological conditions. Briefly, mitochondrial calpain 1 (mit-CPN1) is located within the intermembrane space and mitochondrial matrix. Activation of the mit-CPN1 within intermembrane space cleaves apoptosis inducing factor (AIF), whereas the activated mit-CPN1 within matrix cleaves complex I subunits and metabolic enzymes. Inhibition of the mit-CPN1 could be a potential strategy to decrease cardiac injury during ischemia-reperfusion.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine (Division of Cardiology, Pauley Heart Center), Virginia Commonwealth University, Richmond, VA 23298, United States.
| | - Edward J Lesnefsky
- Department of Medicine (Division of Cardiology, Pauley Heart Center), Virginia Commonwealth University, Richmond, VA 23298, United States; Department of Biochemistry, Virginia Commonwealth University, Richmond, VA 23298, United States; Department of Physiology, Virginia Commonwealth University, Richmond, VA 23298, United States; McGuire VA Medical Center, Richmond, VA 23249, United States
| |
Collapse
|
45
|
Zheng D, Ma J, Yu Y, Li M, Ni R, Wang G, Chen R, Li J, Fan GC, Lacefield JC, Peng T. Silencing of miR-195 reduces diabetic cardiomyopathy in C57BL/6 mice. Diabetologia 2015; 58:1949-58. [PMID: 25994075 PMCID: PMC4499474 DOI: 10.1007/s00125-015-3622-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS MicroRNAs (miRs) have been suggested as potential therapeutic targets for heart diseases. Inhibition of miR-195 prevents apoptosis in cardiomyocytes stimulated with palmitate and transgenic overexpression of miR-195 induces cardiac hypertrophy and heart failure. We investigated whether silencing of miR-195 reduces diabetic cardiomyopathy in a mouse model of streptozotocin (STZ)-induced type 1 diabetes. METHODS Type 1 diabetes was induced in C57BL/6 mice (male, 2 months old) by injections of STZ. RESULTS MiR-195 expression was increased and levels of its target proteins (B cell leukaemia/lymphoma 2 and sirtuin 1) were decreased in STZ-induced type 1 and db/db type 2 diabetic mouse hearts. Systemically delivering an anti-miR-195 construct knocked down miR-195 expression in the heart, reduced caspase-3 activity, decreased oxidative stress, attenuated myocardial hypertrophy and improved myocardial function in STZ-induced mice with a concurrent upregulation of B cell leukaemia/lymphoma 2 and sirtuin 1. Diabetes reduced myocardial capillary density and decreased maximal coronary blood flow in mice. Knockdown of miR-195 increased myocardial capillary density and improved maximal coronary blood flow in diabetic mice. Upregulation of miR-195 sufficiently induced apoptosis in cardiomyocytes and attenuated the angiogenesis of cardiac endothelial cells in vitro. Furthermore, inhibition of miR-195 prevented apoptosis in cardiac endothelial cells in response to NEFA, an important feature of diabetes. CONCLUSIONS/INTERPRETATION Therapeutic silencing of miR-195 reduces myocardial hypertrophy and improves coronary blood flow and myocardial function in diabetes, at least in part by reducing oxidative damage, inhibiting apoptosis and promoting angiogenesis. Thus, miR-195 may represent an alternative therapeutic target for diabetic heart diseases.
Collapse
Affiliation(s)
- Dong Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China 215123
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Jian Ma
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Yong Yu
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Minghui Li
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Rui Ni
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Grace Wang
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Ruizhen Chen
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James C. Lacefield
- Electrical and Computer Engineering, Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China 215123
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
46
|
Nishida K, Yamaguchi O, Otsu K. Degradation systems in heart failure. J Mol Cell Cardiol 2015; 84:212-22. [PMID: 25981331 DOI: 10.1016/j.yjmcc.2015.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 11/29/2022]
Abstract
Heart failure is a complex clinical syndrome that results from any structural or functional impairment of ventricular filling or the ejection of blood, and is a leading cause of morbidity and mortality in industrialized countries. The mechanisms underlying the development of heart failure are multiple, complex and not well understood. Cardiac mass and its homeostasis are maintained by the balance between protein synthesis and degradation, and an imbalance is likely to result in cellular dysfunction and disease. The protein degradation systems are the principle mechanisms for maintaining cellular homeostasis via protein quality control. Three major protein degradation systems have been identified, namely the calpain system, autophagy, and the ubiquitin proteasome system. Proinflammatory mediators involve the development and progression of heart failure. DNA and RNA degradation systems play a critical role in regulating inflammation and maintaining cellular homeostasis mediated by damaged DNA clearance and posttranscriptional regulation, respectively. This review discusses some recent advances in understanding the role of these degradation systems in heart failure.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK.
| |
Collapse
|
47
|
Wan N, Liu X, Zhang XJ, Zhao Y, Hu G, Wan F, Zhang R, Zhu X, Xia H, Li H. Toll-interacting protein contributes to mortality following myocardial infarction through promoting inflammation and apoptosis. Br J Pharmacol 2015; 172:3383-96. [PMID: 25765712 DOI: 10.1111/bph.13130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/27/2015] [Accepted: 03/03/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Toll-interacting protein (Tollip) is an endogenous inhibitor of toll-like receptors, a superfamily that plays a pivotal role in various pathological conditions, including myocardial infarction (MI). However, the exact role of Tollip in MI remains unknown. EXPERIMENTAL APPROACH MI models were established in Tollip knockout (KO) mice, mice with cardiac-specific overexpression of human Tollip gene and in their Tollip(+/+) and non-transgenic controls respectively. The effects of Tollip on MI were evaluated by mortality, infarct size and cardiac function. Hypoxia-induced cardiomyocyte damage was investigated in vitro to confirm the role of Tollip in heart damage. KEY RESULTS Tollip expression was dramatically up-regulated in human ischaemic hearts and infarcted mice hearts. MI-induced mortality, infarct size and cardiac dysfunction were decreased in Tollip-KO mice compared with Tollip(+/+) controls. Ischaemic hearts from Tollip-KO mice exhibited decreased inflammatory cell infiltration and reduced NF-κB activation. Tollip depletion also alleviated myocardial apoptosis by down-regulating pro-apoptotic protein levels and up-regulating anti-apoptotic protein expressions in infarct border zone. Conversely, MI effects were exacerbated in mice with cardiac-specific Tollip overexpression. This aggravated MI injury by Tollip in vivo was confirmed with in vitro assays. Inhibition of Akt signalling was associated with the detrimental effects of Tollip on MI injury; activation of Akt largely reversed the deleterious effects of Tollip on MI-induced cardiomyocyte death. CONCLUSIONS AND IMPLICATIONS Tollip promotes inflammatory and apoptotic responses after MI, leading to increased mortality and aggravated cardiac dysfunction. These findings suggest that Tollip may serve as a novel therapeutic target for the treatment of MI.
Collapse
Affiliation(s)
- Nian Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yichao Zhao
- Department of Cardiology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Gangying Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Fengwei Wan
- Department of Emergency, The Second Artillery General Hospital of Chinese People's Liberation Army Qinghe Clinic, Beijing, China
| | - Rui Zhang
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xueyong Zhu
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Ye T, Wang Q, Zhang Y, Song X, Yang D, Li D, Li D, Su L, Yang Y, Ma S. Over-expression of calpastatin inhibits calpain activation and attenuates post-infarction myocardial remodeling. PLoS One 2015; 10:e0120178. [PMID: 25786109 PMCID: PMC4364764 DOI: 10.1371/journal.pone.0120178] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/21/2015] [Indexed: 11/19/2022] Open
Abstract
Background Calpain is activated following myocardial infarction and ablation of calpastatin (CAST), an endogenous inhibitor of calpains, promotes left ventricular remodeling after myocardial infarction (MI). The present study aimed to investigate the effect of transgenic over-expression of CAST on the post-infarction myocardial remodeling process. Method We established transgenic mice (TG) ubiquitously over-expressing human CAST protein and produced MI in TG mice and C57BL/6J wild-type (WT) littermates. Results The CAST protein expression was profoundly upregulated in the myocardial tissue of TG mice compared with WT littermates (P < 0.01). Overexpression of CAST significantly reduced the infarct size (P < 0.01) and blunted MI-induced interventricular hypertrophy, global myocardial fibrosis and collagen I and collagen III deposition, hypotension and hemodynamic disturbances at 21 days after MI. Moreover, the MI-induced up-regulation and activation of calpains were obviously attenuated in CAST TG mice. MI-induced down-regulation of CAST was partially reversed in TG mice. Additionally, the MI-caused imbalance of matrix metalloproteinases and their inhibitors was improved in TG mice. Conclusions Transgenic over-expression of CAST inhibits calpain activation and attenuates post-infarction myocardial remodeling.
Collapse
Affiliation(s)
- Tingqiao Ye
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Qiang Wang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Yan Zhang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Xiaofeng Song
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Dachun Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - De Li
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Dan Li
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Linan Su
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Yongjian Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
- * E-mail: (YY); (SM)
| | - Shuangtao Ma
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, China
- * E-mail: (YY); (SM)
| |
Collapse
|
49
|
Zheng D, Wang G, Li S, Fan GC, Peng T. Calpain-1 induces endoplasmic reticulum stress in promoting cardiomyocyte apoptosis following hypoxia/reoxygenation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:882-92. [PMID: 25660447 DOI: 10.1016/j.bbadis.2015.01.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/09/2015] [Accepted: 01/31/2015] [Indexed: 12/24/2022]
Abstract
Both calpain activation and endoplasmic reticulum (ER) stress are implicated in ischemic heart injury. However, the role of calpain in ER stress remains largely elusive. This study investigated whether calpain activation causes ER stress, thereby mediating cardiomyocyte apoptosis in an in vitro model of hypoxia/re-oxygenation (H/R). In neonatal mouse cardiomyocytes and rat cardiomyocyte-like H9c2 cells, up-regulation of calpain-1 sufficiently induced ER stress, c-Jun N-terminal protein kinase1/2 (JNK1/2) activation and apoptosis. Inhibition of ER stress or JNK1/2 prevented apoptosis induced by calpain-1. In an in vitro model of H/R-induced injury in cardiomyocytes, H/R was induced by a 24-hour hypoxia followed by a 24-hour re-oxygenation. H/R activated calpain-1, induced ER stress and JNK1/2 activation, and triggered apoptosis. Inhibition of calpain and ER stress blocked JNK1/2 activation and prevented H/R-induced apoptosis. Furthermore, blockade of JNK1/2 signaling inhibited apoptosis following H/R. The role of calpain in ER stress was also demonstrated in an in vivo model of ischemia/reperfusion using transgenic mice over-expressing calpastatin. In summary, calpain-1 induces ER stress and JNK1/2 activation, thereby mediating apoptosis in cardiomyocytes. Accordingly, inhibition of calpain prevents ER stress, JNK1/2 activation and apoptosis in H/R-induced cardiomyocytes. Thus, ER stress/JNK1/2 activation may represent an important mechanism linking calpain-1 to ischemic injury.
Collapse
Affiliation(s)
- Dong Zheng
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China; Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Institute of Cardiovascular Science, Soochow University, Suzhou 215008, China
| | - Grace Wang
- Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada
| | - Shuai Li
- Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati 45267, OH, USA
| | - Tianqing Peng
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China; Critical Illness Research, Lawson Health Research Institute, Canada; Department of Medicine, University of Western Ontario, London, Ontario N6A 4G5, Canada; Department of Pathology, University of Western Ontario, London, Ontario N6A 4G5, Canada; Institute of Cardiovascular Science, Soochow University, Suzhou 215008, China.
| |
Collapse
|
50
|
Neuhof C, Neuhof H. Calpain system and its involvement in myocardial ischemia and reperfusion injury. World J Cardiol 2014; 6:638-652. [PMID: 25068024 PMCID: PMC4110612 DOI: 10.4330/wjc.v6.i7.638] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 01/26/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Calpains are ubiquitous non-lysosomal Ca2+-dependent cysteine proteases also present in myocardial cytosol and mitochondria. Numerous experimental studies reveal an essential role of the calpain system in myocardial injury during ischemia, reperfusion and postischemic structural remodelling. The increasing Ca2+-content and Ca2+-overload in myocardial cytosol and mitochondria during ischemia and reperfusion causes an activation of calpains. Upon activation they are able to injure the contractile apparatus and impair the energy production by cleaving structural and functional proteins of myocytes and mitochondria. Besides their causal involvement in acute myocardial dysfunction they are also involved in structural remodelling after myocardial infarction by the generation and release of proapoptotic factors from mitochondria. Calpain inhibition can prevent or attenuate myocardial injury during ischemia, reperfusion, and in later stages of myocardial infarction.
Collapse
|