1
|
Patnaik S, Nathan S, Kar B, Gregoric ID, Li YP. The Role of Extracellular Heat Shock Proteins in Cardiovascular Diseases. Biomedicines 2023; 11:1557. [PMID: 37371652 DOI: 10.3390/biomedicines11061557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
In the early 1960s, heat shock proteins (HSPs) were first identified as vital intracellular proteinaceous components that help in stress physiology and reprogram the cellular responses to enable the organism's survival. By the early 1990s, HSPs were detected in extracellular spaces and found to activate gamma-delta T-lymphocytes. Subsequent investigations identified their association with varied disease conditions, including autoimmune disorders, diabetes, cancer, hepatic, pancreatic, and renal disorders, and cachexia. In cardiology, extracellular HSPs play a definite, but still unclear, role in atherosclerosis, acute coronary syndromes, and heart failure. The possibility of HSP-targeted novel molecular therapeutics has generated much interest and hope in recent years. In this review, we discuss the role of Extracellular Heat Shock Proteins (Ec-HSPs) in various disease states, with a particular focus on cardiovascular diseases.
Collapse
Affiliation(s)
- Soumya Patnaik
- Division of Cardiology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sriram Nathan
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Biswajit Kar
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Igor D Gregoric
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yi-Ping Li
- Division of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
2
|
Papini G, Furini G, Matteucci M, Biemmi V, Casieri V, Di Lascio N, Milano G, Chincoli LR, Faita F, Barile L, Lionetti V. Cardiomyocyte-targeting exosomes from sulforaphane-treated fibroblasts affords cardioprotection in infarcted rats. J Transl Med 2023; 21:313. [PMID: 37161563 PMCID: PMC10169450 DOI: 10.1186/s12967-023-04155-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Exosomes (EXOs), tiny extracellular vesicles that facilitate cell-cell communication, are being explored as a heart failure treatment, although the features of the cell source restrict their efficacy. Fibroblasts the most prevalent non-myocyte heart cells, release poor cardioprotective EXOs. A noninvasive method for manufacturing fibroblast-derived exosomes (F-EXOs) that target cardiomyocytes and slow cardiac remodeling is expected. As a cardioprotective isothiocyanate, sulforaphane (SFN)-induced F-EXOs (SFN-F-EXOs) should recapitulate its anti-remodeling properties. METHODS Exosomes from low-dose SFN (3 μM/7 days)-treated NIH/3T3 murine cells were examined for number, size, and protein composition. Fluorescence microscopy, RT-qPCR, and western blot assessed cell size, oxidative stress, AcH4 levels, hypertrophic gene expression, and caspase-3 activation in angiotensin II (AngII)-stressed HL-1 murine cardiomyocytes 12 h-treated with various EXOs. The uptake of fluorescently-labeled EXOs was also measured in cardiomyocytes. The cardiac function of infarcted male Wistar rats intramyocardially injected with different EXOs (1·1012) was examined by echocardiography. Left ventricular infarct size, hypertrophy, and capillary density were measured. RESULTS Sustained treatment of NIH/3T3 with non-toxic SFN concentration significantly enhances the release of CD81 + EXOs rich in TSG101 (Tumor susceptibility gene 101) and Hsp70 (Heat Shock Protein 70), and containing maspin, an endogenous histone deacetylase 1 inhibitor. SFN-F-EXOs counteract angiotensin II (AngII)-induced hypertrophy and apoptosis in murine HL-1 cardiomyocytes enhancing SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a) levels more effectively than F-EXOs. In stressed cardiomyocytes, SFN-F-EXOs boost AcH4 levels by 30% (p < 0.05) and significantly reduce oxidative stress more than F-EXOs. Fluorescence microscopy showed that mouse cardiomyocytes take in SFN-F-EXOs ~ threefold more than F-EXOs. Compared to vehicle-injected infarcted hearts, SFN-F-EXOs reduce hypertrophy, scar size, and improve contractility. CONCLUSIONS Long-term low-dose SFN treatment of fibroblasts enhances the release of anti-remodeling cardiomyocyte-targeted F-EXOs, which effectively prevent the onset of HF. The proposed method opens a new avenue for large-scale production of cardioprotective exosomes for clinical application using allogeneic fibroblasts.
Collapse
Affiliation(s)
- Gaia Papini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giulia Furini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy
| | - Marco Matteucci
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Vanessa Biemmi
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Valentina Casieri
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Nicole Di Lascio
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giuseppina Milano
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lucia Rosa Chincoli
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Lucio Barile
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy.
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy.
| |
Collapse
|
3
|
Yu Q, Ju P, Kou W, Zhai M, Zeng Y, Maimaitiaili N, Shi Y, Xu X, Zhao Y, Jian W, Feinberg MW, Xu Y, Zhuang J, Peng W. Macrophage-Specific NLRC5 Protects From Cardiac Remodeling Through Interaction With HSPA8. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
4
|
Zhang Y, Wu J, Dong E, Wang Z, Xiao H. Toll-like receptors in cardiac hypertrophy. Front Cardiovasc Med 2023; 10:1143583. [PMID: 37113698 PMCID: PMC10126280 DOI: 10.3389/fcvm.2023.1143583] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that can identify pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). TLRs play an important role in the innate immune response, leading to acute and chronic inflammation. Cardiac hypertrophy, an important cardiac remodeling phenotype during cardiovascular disease, contributes to the development of heart failure. In previous decades, many studies have reported that TLR-mediated inflammation was involved in the induction of myocardium hypertrophic remodeling, suggesting that targeting TLR signaling might be an effective strategy against pathological cardiac hypertrophy. Thus, it is necessary to study the mechanisms underlying TLR functions in cardiac hypertrophy. In this review, we summarized key findings of TLR signaling in cardiac hypertrophy.
Collapse
Affiliation(s)
- Yanan Zhang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Correspondence: Zhanli Wang Han Xiao
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Correspondence: Zhanli Wang Han Xiao
| |
Collapse
|
5
|
de Oliveira AA, Mendoza VO, Rastogi S, Nunes KP. New insights into the role and therapeutic potential of HSP70 in diabetes. Pharmacol Res 2022; 178:106173. [PMID: 35278625 DOI: 10.1016/j.phrs.2022.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
Abstract
Emerging evidence indicates that HSP70 represents a key mechanism in the pathophysiology of β-cell dysfunction, insulin resistance, and various diabetic complications, including micro- and macro-vascular alterations, as well as impaired hemostasis. Hyperglycemia, a hallmark of both types of diabetes, increases the circulating levels of HSP70 (eHSP70), but there is still divergence about whether diabetes up- or down-regulates the intracellular fraction of this protein (iHSP70). Here, we consider that iHSP70 levels reduce in diabetic arterial structures and that the vascular system is in direct contact with all other systems in the body suggesting that a systemic response might also be happening for iHSP70, which is characterized by decreased levels of HSP70 in the vasculature. Furthermore, although many pathways have been proposed to explain HSP70's functions in diabetes, and organs/tissues/cells-specific variations occur, the membrane-bound receptor of the innate immune system, Toll-like receptor 4, and its downstream signal transduction pathways appear to be a constant, not only when we explore the actions of eHSP70, but also when we assess the contributions of iHSP70. In this review, we focus on discussing the multiple roles of HSP70 across organs/tissues/cells affected by hyperglycemia to further explore the possibility of targeting this protein with pharmacological and non-pharmacological approaches in the context of diabetes.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Valentina Ochoa Mendoza
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Swasti Rastogi
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States.
| |
Collapse
|
6
|
Wang Y, Wu J, Wang D, Yang R, Liu Q. Traditional Chinese Medicine Targeting Heat Shock Proteins as Therapeutic Strategy for Heart Failure. Front Pharmacol 2022; 12:814243. [PMID: 35115946 PMCID: PMC8804377 DOI: 10.3389/fphar.2021.814243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) is the terminal stage of multifarious heart diseases and is responsible for high hospitalization rates and mortality. Pathophysiological mechanisms of HF include cardiac hypertrophy, remodeling and fibrosis resulting from cell death, inflammation and oxidative stress. Heat shock proteins (HSPs) can ameliorate folding of proteins, maintain protein structure and stability upon stress, protect the heart from cardiac dysfunction and ameliorate apoptosis. Traditional Chinese medicine (TCM) regulates expression of HSPs and has beneficial therapeutic effect in HF. In this review, we summarized the function of HSPs in HF and the role of TCM in regulating expression of HSPs. Studying the regulation of HSPs by TCM will provide novel ideas for the study of the mechanism and treatment of HF.
Collapse
Affiliation(s)
- Yanchun Wang
- Shenyang the Tenth People’s Hospital, Shenyang, China
| | - Junxuan Wu
- Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Dawei Wang
- Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| | - Rongyuan Yang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| | - Qing Liu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, China
- *Correspondence: Qing Liu, ; Dawei Wang, ; Rongyuan Yang,
| |
Collapse
|
7
|
Gerbino A, Forleo C, Milano S, Piccapane F, Procino G, Pepe M, Piccolo M, Guida P, Resta N, Favale S, Svelto M, Carmosino M. Pro-inflammatory cytokines as emerging molecular determinants in cardiolaminopathies. J Cell Mol Med 2021; 25:10902-10915. [PMID: 34773379 PMCID: PMC8642682 DOI: 10.1111/jcmm.16975] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Mutations in Lamin A/C gene (lmna) cause a wide spectrum of cardiolaminopathies strictly associated with significant deterioration of the electrical and contractile function of the heart. Despite the continuous flow of biomedical evidence, linking cardiac inflammation to heart remodelling in patients harbouring lmna mutations is puzzling. Therefore, we profiled 30 serum cytokines/chemokines in patients belonging to four different families carrying pathogenic lmna mutations segregating with cardiac phenotypes at different stages of severity (n = 19) and in healthy subjects (n = 11). Regardless lmna mutation subtype, high levels of circulating granulocyte colony‐stimulating factor (G‐CSF) and interleukin 6 (IL‐6) were found in all affected patients’ sera. In addition, elevated levels of Interleukins (IL) IL‐1Ra, IL‐1β IL‐4, IL‐5 and IL‐8 and the granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) were measured in a large subset of patients associated with more aggressive clinical manifestations. Finally, the expression of the pro‐inflammatory 70 kDa heat shock protein (Hsp70) was significantly increased in serum exosomes of patients harbouring the lmna mutation associated with the more severe phenotype. Overall, the identification of patient subsets with overactive or dysregulated myocardial inflammatory responses could represent an innovative diagnostic, prognostic and therapeutic tool against Lamin A/C cardiomyopathies.
Collapse
Affiliation(s)
- Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Cinzia Forleo
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Francesca Piccapane
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Martino Pepe
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Mara Piccolo
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Piero Guida
- Regional General Hospital "F. Miulli", Acquaviva delle Fonti, Italy
| | - Nicoletta Resta
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Stefano Favale
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
8
|
Junho CVC, Azevedo CAB, da Cunha RS, de Yurre AR, Medei E, Stinghen AEM, Carneiro-Ramos MS. Heat Shock Proteins: Connectors between Heart and Kidney. Cells 2021; 10:cells10081939. [PMID: 34440708 PMCID: PMC8391307 DOI: 10.3390/cells10081939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022] Open
Abstract
Over the development of eukaryotic cells, intrinsic mechanisms have been developed in order to provide the ability to defend against aggressive agents. In this sense, a group of proteins plays a crucial role in controlling the production of several proteins, guaranteeing cell survival. The heat shock proteins (HSPs), are a family of proteins that have been linked to different cellular functions, being activated under conditions of cellular stress, not only imposed by thermal variation but also toxins, radiation, infectious agents, hypoxia, etc. Regarding pathological situations as seen in cardiorenal syndrome (CRS), HSPs have been shown to be important mediators involved in the control of gene transcription and intracellular signaling, in addition to be an important connector with the immune system. CRS is classified as acute or chronic and according to the first organ to suffer the injury, which can be the heart (CRS type 1 and type 2), kidneys (CRS type 3 and 4) or both (CRS type 5). In all types of CRS, the immune system, redox balance, mitochondrial dysfunction, and tissue remodeling have been the subject of numerous studies in the literature in order to elucidate mechanisms and propose new therapeutic strategies. In this sense, HSPs have been targeted by researchers as important connectors between kidney and heart. Thus, the present review has a focus to present the state of the art regarding the role of HSPs in the pathophysiology of cardiac and renal alterations, as well their role in the kidney–heart axis.
Collapse
Affiliation(s)
- Carolina Victória Cruz Junho
- Center of Natural and Human Sciences (CCNH), Laboratory of Cardiovascular Immunology, Federal University of ABC, Santo André 09210-580, Brazil
| | - Carolina Amaral Bueno Azevedo
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil
| | - Regiane Stafim da Cunha
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil
| | - Ainhoa Rodriguez de Yurre
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Emiliano Medei
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro 21941-902, Brazil
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 22281-100, Brazil
| | - Andréa Emilia Marques Stinghen
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil
| | - Marcela Sorelli Carneiro-Ramos
- Center of Natural and Human Sciences (CCNH), Laboratory of Cardiovascular Immunology, Federal University of ABC, Santo André 09210-580, Brazil
| |
Collapse
|
9
|
Wei XH, Guo X, Pan CS, Li H, Cui YC, Yan L, Fan JY, Deng JN, Hu BH, Chang X, He SY, Yan LL, Sun K, Wang CS, Han JY. Quantitative Proteomics Reveal That Metabolic Improvement Contributes to the Cardioprotective Effect of T 89 on Isoproterenol-Induced Cardiac Injury. Front Physiol 2021; 12:653349. [PMID: 34262469 PMCID: PMC8273540 DOI: 10.3389/fphys.2021.653349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/12/2021] [Indexed: 02/03/2023] Open
Abstract
Background T89, a traditional Chinese medicine, has passed phase II, and is undergoing phase III clinical trials for treatment of ischemic cardiovascular disease by the US FDA. However, the role of T89 on isoproterenol (ISO)-induced cardiac injury is unknown. The present study aimed to explore the effect and underlying mechanism of T89 on ISO-induced cardiac injury. Methods Male Sprague-Dawley rats received subcutaneous injection of ISO saline solution at 24 h intervals for the first 3 days and then at 48 h intervals for the next 12 days. T89 at dose of 111.6 and 167.4 mg/kg was administrated by gavage for 15 consecutive days. Rat survival rate, cardiac function evaluation, morphological observation, quantitative proteomics, and Western blotting analysis were performed. Results T89 obviously improved ISO-induced low survival rate, attenuated ISO-evoked cardiac injury, as evidenced by myocardial blood flow, heart function, and morphology. Quantitative proteomics revealed that the cardioprotective effect of T89 relied on the regulation of metabolic pathways, including glycolipid metabolism and energy metabolism. T89 inhibited the enhancement of glycolysis, promoted fatty acid oxidation, and restored mitochondrial oxidative phosphorylation by regulating Eno1, Mcee, Bdh1, Ces1c, Apoc2, Decr1, Acaa2, Cbr4, ND2, Cox 6a, Cox17, ATP5g, and ATP5j, thus alleviated oxidative stress and energy metabolism disorder and ameliorated cardiac injury after ISO. The present study also verified that T89 significantly restrained ISO-induced increase of HSP70/HSP40 and suppressed the phosphorylation of ERK, further restored the expression of CX43, confirming the protective role of T89 in cardiac hypertrophy. Proteomics data are available via ProteomeXchange with identifier PXD024641. Conclusion T89 reduced mortality and improves outcome in the model of ISO-induced cardiac injury and the cardioprotective role of T89 is correlated with the regulation of glycolipid metabolism, recovery of mitochondrial function, and improvement of myocardial energy.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Xiao Guo
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Huan Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Yuan-Chen Cui
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Jing-Na Deng
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Bai-He Hu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Xin Chang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Shu-Ya He
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Lu-Lu Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Kai Sun
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Chuan-She Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Hamdani N, Costantino S, Mügge A, Lebeche D, Tschöpe C, Thum T, Paneni F. Leveraging clinical epigenetics in heart failure with preserved ejection fraction: a call for individualized therapies. Eur Heart J 2021; 42:1940-1958. [PMID: 36282124 DOI: 10.1093/eurheartj/ehab197] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/17/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Described as the 'single largest unmet need in cardiovascular medicine', heart failure with preserved ejection fraction (HFpEF) remains an untreatable disease currently representing 65% of new heart failure diagnoses. HFpEF is more frequent among women and associates with a poor prognosis and unsustainable healthcare costs. Moreover, the variability in HFpEF phenotypes amplifies complexity and difficulties in the approach. In this perspective, unveiling novel molecular targets is imperative. Epigenetic modifications-defined as changes of DNA, histones, and non-coding RNAs (ncRNAs)-represent a molecular framework through which the environment modulates gene expression. Epigenetic signals acquired over the lifetime lead to chromatin remodelling and affect transcriptional programmes underlying oxidative stress, inflammation, dysmetabolism, and maladaptive left ventricular remodelling, all conditions predisposing to HFpEF. The strong involvement of epigenetic signalling in this setting makes the epigenetic information relevant for diagnostic and therapeutic purposes in patients with HFpEF. The recent advances in high-throughput sequencing, computational epigenetics, and machine learning have enabled the identification of reliable epigenetic biomarkers in cardiovascular patients. Contrary to genetic tools, epigenetic biomarkers mirror the contribution of environmental cues and lifestyle changes and their reversible nature offers a promising opportunity to monitor disease states. The growing understanding of chromatin and ncRNAs biology has led to the development of several Food and Drug Administration approved 'epidrugs' (chromatin modifiers, mimics, anti-miRs) able to prevent transcriptional alterations underpinning left ventricular remodelling and HFpEF. In the present review, we discuss the importance of clinical epigenetics as a new tool to be employed for a personalized management of HFpEF.
Collapse
Affiliation(s)
- Nazha Hamdani
- Institute of Physiology, Ruhr University, Bochum, Germany.,Molecular and Experimental Cardiology, Ruhr University, Bochum, Germany.,Department of Cardiology, St-Josef Hospital, Ruhr University, Bochum, Germany.,Clinical Pharmacology, Ruhr University, Bochum, Germany
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland
| | - Andreas Mügge
- Molecular and Experimental Cardiology, Ruhr University, Bochum, Germany.,Department of Cardiology, St-Josef Hospital, Ruhr University, Bochum, Germany
| | - Djamel Lebeche
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY 10029, USA.,Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Medicine, Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover 30625, Germany
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zürich, Switzerland
| |
Collapse
|
11
|
de Oliveira AA, Priviero F, Webb RC, Nunes KP. Impaired HSP70 Expression in the Aorta of Female Rats: A Novel Insight Into Sex-Specific Differences in Vascular Function. Front Physiol 2021; 12:666696. [PMID: 33967836 PMCID: PMC8100344 DOI: 10.3389/fphys.2021.666696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/25/2021] [Indexed: 12/19/2022] Open
Abstract
Heat-shock protein 70 (HSP70) contributes to cellular calcium (Ca2+) handling mechanisms during receptor-mediated vascular contraction. Interestingly, previous studies have independently reported sex-related differences in HSP70 expression and Ca2+ dynamics. Still, it is unknown if sex, as a variable, plays a role in the impact that HSP70 has upon vascular contraction. To narrow this gap, we investigated if differences exist in the expression levels of HSP70 in the aorta, and if targeting this protein contributes to sex disparity in vascular responses. We report that, compared with male animals, female rats present a reduction in the basal levels of HSP70. More compelling, we found that the blockade of HSP70 has a greater impact on phenylephrine-induced phasic and tonic vascular contraction in female animals. In fact, it seems that the inhibition of HSP70 significantly affects vascular Ca2+ handling mechanisms in females, which could be associated with the fact that these animals have impaired HSP70 expression. Corroborating this idea, we uncovered that the higher sensitivity of female rats to HSP70 inhibition does not involve an increase in NO-dependent vasodilation nor a decrease in vascular oxidative stress. In summary, our findings reveal a novel mechanism associated with sex-specific differences in vascular responses to α-1 adrenergic stimulation, which might contribute to unraveling the network of intertwined pathways conferring female protection to (cardio)vascular diseases.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Fernanda Priviero
- Department of Physiology, Augusta University, Augusta, GA, United States
- Department of Cell Biology and Anatomy, Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - R. Clinton Webb
- Department of Cell Biology and Anatomy, Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| |
Collapse
|
12
|
de Oliveira AA, Priviero F, Tostes RC, Webb RC, Nunes KP. Dissecting the interaction between HSP70 and vascular contraction: role of [Formula: see text] handling mechanisms. Sci Rep 2021; 11:1420. [PMID: 33446873 PMCID: PMC7809064 DOI: 10.1038/s41598-021-80966-6] [Citation(s) in RCA: 9] [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: 10/05/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023] Open
Abstract
Heat-shock protein 70 (HSP70) is a ubiquitously expressed molecular chaperone with various biological functions. Recently, we demonstrated that HSP70 is key for adequate vascular reactivity. However, the specific mechanisms targeted by HSP70 to assist in this process remain elusive. Since there is a wealth of evidence connecting HSP70 to calcium ([Formula: see text]), a master regulator of contraction, we designed this study to investigate whether blockade of HSP70 disrupts vascular contraction via impairment of [Formula: see text] handling mechanisms. We performed functional studies in aortas isolated from male Sprague Dawley rats in the presence or absence of exogenous [Formula: see text], and we determined the effects of VER155008, an inhibitor of HSP70, on [Formula: see text] handling as well as key mechanisms that regulate vascular contraction. Changes in the intracellular concentration of [Formula: see text] were measured with a biochemical assay kit. We report that blockade of HSP70 leads to [Formula: see text] mishandling in aorta stimulated with phenylephrine, decreasing both phasic and tonic contractions. Importantly, in [Formula: see text] free Krebs' solution, inhibition of HSP70 only reduced the [Formula: see text] of the phasic contraction if the protein was blocked before IP3r-mediated [Formula: see text] release, suggesting that HSP70 has a positive effect towards this receptor. Corroborating this statement, VER155008 did not potentiate an IP3r inhibitor's outcomes, even with partial blockade. In another set of experiments, the inhibition of HSP70 attenuated the amplitude of the tonic contraction independently of the moment VER155008 was added to the chamber (i.e., whether it was before or after IP3r-mediated phasic contraction). More compelling, following re-addition of [Formula: see text], VER155008 amplified the inhibitory effects of a voltage-dependent [Formula: see text] channel blocker, but not of a voltage-independent [Formula: see text] channel inhibitor, indicating that HSP70 has a positive impact on the latter. Lastly, the mechanism by which HSP70 modulates vascular contraction does not involve the [Formula: see text] sensitizer protein, Rho-kinase, nor the SERCA pump, as blockade of these proteins in the presence of VER155008 almost abolished contraction. In summary, our findings shed light on the processes targeted by HSP70 during vascular contraction and open research avenues for potential new mechanisms in vascular diseases.
Collapse
Affiliation(s)
- Amanda A. de Oliveira
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, USA
| | - Fernanda Priviero
- Department of Physiology, Augusta University, Augusta, USA
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, USA
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - R. Clinton Webb
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, USA
| | - Kenia P. Nunes
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, USA
| |
Collapse
|
13
|
Xiao Z, Kong B, Yang H, Dai C, Fang J, Qin T, Huang H. Key Player in Cardiac Hypertrophy, Emphasizing the Role of Toll-Like Receptor 4. Front Cardiovasc Med 2020; 7:579036. [PMID: 33324685 PMCID: PMC7725871 DOI: 10.3389/fcvm.2020.579036] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptor 4 (TLR4), a key pattern recognition receptor, initiates the innate immune response and leads to chronic and acute inflammation. In the past decades, accumulating evidence has implicated TLR4-mediated inflammatory response in regulation of myocardium hypertrophic remodeling, indicating that regulation of the TLR4 signaling pathway may be an effective strategy for managing cardiac hypertrophy's pathophysiology. Given TLR4's significance, it is imperative to review the molecular mechanisms and roles underlying TLR4 signaling in cardiac hypertrophy. Here, we comprehensively review the current knowledge of TLR4-mediated inflammatory response and its interaction ligands and co-receptors, as well as activation of various intracellular signaling. We also describe the associated roles in promoting immune cell infiltration and inflammatory mediator secretion, that ultimately cause cardiac hypertrophy. Finally, we provide examples of some of the most promising drugs and new technologies that have the potential to attenuate TLR4-mediated inflammatory response and prevent or reverse the ominous cardiac hypertrophy outcomes.
Collapse
Affiliation(s)
- Zheng Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chang Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jin Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Tianyou Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
14
|
Seclì L, Sorge M, Morotti A, Brancaccio M. Blocking Extracellular Chaperones to Improve Cardiac Regeneration. Front Bioeng Biotechnol 2020; 8:411. [PMID: 32528937 PMCID: PMC7264090 DOI: 10.3389/fbioe.2020.00411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/14/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic or acute insults to the myocardium are responsible for the onset of cardiomyopathy and heart failure. Due to the poor regenerative ability of the human adult heart, the survival of cardiomyocytes is a prerequisite to support heart function. Chaperone proteins, by regulating sarcomeric protein folding, function, and turnover in the challenging environment of the beating heart, play a fundamental role in myocardial physiology. Nevertheless, a number of evidences indicate that, under stress conditions or during cell damage, myocardial cells release chaperone proteins that, from the extracellular milieu, play a detrimental function, by perpetuating inflammation and inducing cardiomyocyte apoptosis. Blocking the activity of extracellular chaperones has been proven to have beneficial effects on heart function in preclinical models of myocardial infarction and cardiomyopathy. The application of this approach in combination with tissue engineering strategies may represent a future innovation in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Matteo Sorge
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
15
|
Atorvastatin Improves Doxorubicin-Induced Cardiac Dysfunction by Modulating Hsp70, Akt, and MAPK Signaling Pathways. J Cardiovasc Pharmacol 2020; 73:223-231. [PMID: 30540688 DOI: 10.1097/fjc.0000000000000646] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Atorvastatin is a lipid-regulating drug that is commonly used in clinical practice and can stabilize plaques. Increasing evidence shows that statins have anti-heart failure (HF) effects, but their specific mechanism is not clear. The purpose of this study was to investigate the cardioprotective effects of atorvastatin on HF in rats and its mechanism. Continuous intraperitoneal injection of 2.5 mg/kg/w doxorubicin for 6 weeks, with a cumulative dose of 15 mg/kg, was used to induce a rat model of HF. Then, the rats were treated with low-dose atorvastatin, high-dose atorvastatin, or saline for 4 weeks. In the DOX-treated groups, echocardiography showed decreases in left ventricular ejection fraction and fractional shortening and increases in left ventricular end-diastolic diameter and left ventricular posterior wall thickness compared with those in the control group, and increased levels of brain natriuretic peptide and Hsp70 were also found in the doxorubicin-treated groups. Compared with saline intervention, atorvastatin ameliorated left ventricular ejection fraction, fractional shortening, left ventricular end-diastolic diameter, and left ventricular posterior wall thickness (a significant difference was observed only in the high-dose group) and reduced serum brain natriuretic peptide. Hematoxylin and eosin staining showed that atorvastatin ameliorated myocardial injury. The improvement in cardiac function induced by atorvastatin was accompanied by increased Hsp70 expression, decreased p-ERK and p-JNK expression, and a reduction in myocardial fibrosis shown by Masson staining. In addition, atorvastatin had a protective effect on the myocardial apoptosis signaling pathway, with increased p-Akt expression and downregulated cleaved caspase-3 expression, and the reduction in myocardial apoptosis was confirmed by a TUNEL assay. Therefore, our experiments demonstrated that atorvastatin may protect cardiac function by modulating Hsp70, p-Akt, p-ERK, and p-JNK signaling to reduce myocardial fibrosis and myocardial apoptosis.
Collapse
|
16
|
Yadav K, Yadav A, Vashistha P, Pandey VP, Dwivedi UN. Protein Misfolding Diseases and Therapeutic Approaches. Curr Protein Pept Sci 2020; 20:1226-1245. [PMID: 31187709 DOI: 10.2174/1389203720666190610092840] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/01/2019] [Accepted: 02/24/2019] [Indexed: 12/12/2022]
Abstract
Protein folding is the process by which a polypeptide chain acquires its functional, native 3D structure. Protein misfolding, on the other hand, is a process in which protein fails to fold into its native functional conformation. This misfolding of proteins may lead to precipitation of a number of serious diseases such as Cystic Fibrosis (CF), Alzheimer's Disease (AD), Parkinson's Disease (PD), and Amyotrophic Lateral Sclerosis (ALS) etc. Protein Quality-control (PQC) systems, consisting of molecular chaperones, proteases and regulatory factors, help in protein folding and prevent its aggregation. At the same time, PQC systems also do sorting and removal of improperly folded polypeptides. Among the major types of PQC systems involved in protein homeostasis are cytosolic, Endoplasmic Reticulum (ER) and mitochondrial ones. The cytosol PQC system includes a large number of component chaperones, such as Nascent-polypeptide-associated Complex (NAC), Hsp40, Hsp70, prefoldin and T Complex Protein-1 (TCP-1) Ring Complex (TRiC). Protein misfolding diseases caused due to defective cytosolic PQC system include diseases involving keratin/collagen proteins, cardiomyopathies, phenylketonuria, PD and ALS. The components of PQC system of Endoplasmic Reticulum (ER) include Binding immunoglobulin Protein (BiP), Calnexin (CNX), Calreticulin (CRT), Glucose-regulated Protein GRP94, the thiol-disulphide oxidoreductases, Protein Disulphide Isomerase (PDI) and ERp57. ER-linked misfolding diseases include CF and Familial Neurohypophyseal Diabetes Insipidus (FNDI). The components of mitochondrial PQC system include mitochondrial chaperones such as the Hsp70, the Hsp60/Hsp10 and a set of proteases having AAA+ domains similar to the proteasome that are situated in the matrix or the inner membrane. Protein misfolding diseases caused due to defective mitochondrial PQC system include medium-chain acyl-CoA dehydrogenase (MCAD)/Short-chain Acyl-CoA Dehydrogenase (SCAD) deficiency diseases, hereditary spastic paraplegia. Among therapeutic approaches towards the treatment of various protein misfolding diseases, chaperones have been suggested as potential therapeutic molecules for target based treatment. Chaperones have been advantageous because of their efficient entry and distribution inside the cells, including specific cellular compartments, in therapeutic concentrations. Based on the chemical nature of the chaperones used for therapeutic purposes, molecular, chemical and pharmacological classes of chaperones have been discussed.
Collapse
Affiliation(s)
- Kusum Yadav
- Department of Biochemistry, University of Lucknow, Lucknow, U.P, India
| | - Anurag Yadav
- Department of Microbiology, College of Basic Sciences and Humanities, Sardar Krushinagar Dantiwada Agricultural University, Banaskantha, Gujarat, India
| | | | - Veda P Pandey
- Department of Biochemistry, University of Lucknow, Lucknow, U.P, India
| | - Upendra N Dwivedi
- Department of Biochemistry, University of Lucknow, Lucknow, U.P, India.,Institute for Development of Advanced Computing, ONGC Centre for Advanced Studies, University of Lucknow, Lucknow, U.P., India
| |
Collapse
|
17
|
Lu X, He Y, Tang C, Wang X, Que L, Zhu G, Liu L, Ha T, Chen Q, Li C, Xu Y, Li J, Li Y. Triad3A attenuates pathological cardiac hypertrophy involving the augmentation of ubiquitination-mediated degradation of TLR4 and TLR9. Basic Res Cardiol 2020; 115:19. [PMID: 32008145 DOI: 10.1007/s00395-020-0779-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Activation of TLRs mediated the NF-κB signaling pathway plays an important pathophysiological role in cardiac hypertrophy. Triad3A, a ubiquitin E3 ligase, has been reported to negatively regulate NF-κB activation pathway via promoting ubiquitination and degradation of TLR4 and TLR9 in innate immune cells. The role of Triad3A in cardiac hypertrophic development remains unknown. The present study investigated whether there is a link between Triad3A and TLR4 and TLR9 in pressure overload induced cardiac hypertrophy. We observed that Triad3A levels were markedly reduced following transverse aortic constriction (TAC) induced cardiac hypertrophy. Similarly, stimulation of neonatal rat cardiac myocytes (NRCMs) with angiotensin-II (Ang II) significantly decreased Triad3A expression. To determine the role of Triad3A in TAC-induced cardiac hypertrophy, we transduced the myocardium with adenovirus expressing Triad3A followed by induction of TAC. We observed that increased expression of Triad3A significantly attenuated cardiac hypertrophy and improved cardiac function. To investigate the mechanisms by which Triad3A attenuated cardiac hypertrophy, we examined the Triad3A E3 ubiquitination on TLR4 and TLR9. We found that Triad3A promoted TLR4 and TLR9 degradation through ubiquitination. Triad3A mediated TLR4 and TLR9 degradation resulted in suppression of NF-κB activation. Our data suggest that Triad3A plays a protective role in the development of cardiac hypertrophy, at least through catalyzing ubiquitination-mediated degradation of TLR4 and TLR9, thus negatively regulating NF-κB activation.
Collapse
Affiliation(s)
- Xia Lu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yijie He
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chao Tang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoyang Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Linli Que
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guoqing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tuanzhu Ha
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN, 37614-0575, USA
| | - Qi Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN, 37614-0575, USA
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Jiantao Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center For Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
18
|
Liu P, Bao HY, Jin CC, Zhou JC, Hua F, Li K, Lv XX, Cui B, Hu ZW, Zhang XW. Targeting Extracellular Heat Shock Protein 70 Ameliorates Doxorubicin-Induced Heart Failure Through Resolution of Toll-Like Receptor 2-Mediated Myocardial Inflammation. J Am Heart Assoc 2019; 8:e012338. [PMID: 31576776 PMCID: PMC6818050 DOI: 10.1161/jaha.119.012338] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Heart failure (HF) is one of the most significant causes of morbidity and mortality for the cardiovascular risk population. We found previously that extracellular HSP70 (heat shock protein) is an important trigger in cardiac hypertrophy and fibrosis, which are associated with the development of heart dysfunction. However, the potential role of HSP70 in response to HF and whether it could be a target for the therapy of HF remain unknown. Methods and Results An HF mouse model was generated by a single IP injection of doxorubicin at a dose of 15 mg/kg. Ten days later, these mice were treated with an HSP70 neutralizing antibody for 5 times. We observed that doxorubicin treatment increased circulating HSP70 and expression of HSP70 in myocardium and promoted its extracellular release in the heart. Blocking extracellular HSP70 activity by its antibody significantly ameliorated doxorubicin‐induced left ventricular dilation and dysfunction, which was accompanied by a significant inhibition of cardiac fibrosis. The cardioprotective effect of the anti‐HSP70 antibody was largely attributed to its ability to promote the resolution of myocardial inflammation, as evidenced by its suppression of the toll‐like receptor 2–associated signaling cascade and modulation of the intracellular distribution of the p50 and p65 subunits of nuclear factor‐κB. Conclusions Extracellular HSP70 serves as a noninfectious inflammatory factor in the development of HF, and blocking extracellular HSP70 activity may provide potential therapeutic benefits for the treatment of HF.
Collapse
Affiliation(s)
- Peng Liu
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Hua-Yan Bao
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Cai-Cai Jin
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Ji-Chao Zhou
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Fang Hua
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Ke Li
- Institute of Medicinal Biotechnology Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xiao-Xi Lv
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Bing Cui
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Zhuo-Wei Hu
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xiao-Wei Zhang
- Molecular Immunology and Pharmacology Group State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
19
|
Wischmeyer PE, Mintz-Cole RA, Baird CH, Easley KA, May AK, Sax HC, Kudsk KA, Hao L, Tran PH, Jones DP, Blumberg HM, Ziegler TR. Role of heat shock protein and cytokine expression as markers of clinical outcomes with glutamine-supplemented parenteral nutrition in surgical ICU patients. Clin Nutr 2019; 39:563-573. [PMID: 30981628 DOI: 10.1016/j.clnu.2019.02.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nutrients, such as glutamine (GLN), have been shown to effect levels of a family of protective proteins termed heat shock proteins (HSPs) in experimental and clinical critical illness. HSPs are believed to serve as extracellular inflammatory messengers and intracellular cytoprotective molecules. Extracellular HSP70 (eHSP70) has been termed a chaperokine due to ability to modulate the immune response. Altered levels of eHSP70 are associated with various disease states. Larger clinical trial data on GLN effect on eHSP expression and eHSP70's association with inflammatory mediators and clinical outcomes in critical illness are limited. OBJECTIVE Explore effect of longitudinal change in serum eHSP70, eHSP27 and inflammatory cytokine levels on clinical outcomes such as pneumonia and mortality in adult surgical intensive care unit (SICU) patients. Further, evaluate effect of parenteral nutrition (PN) supplemented with GLN (GLN-PN) versus GLN-free, standard PN (STD-PN) on serum eHSP70 and eHSP27 concentrations. METHODS Secondary observational analysis of a multicenter clinical trial in 150 adults after cardiac, vascular, or gastrointestinal surgery requiring PN support and SICU care conducted at five academic medical centers. Patients received isocaloric, isonitrogenous PN, with or without GLN dipeptide. Serum eHSP70 and eHSP27, interleukin-6 (IL-6), and 8 (IL-8) concentrations were analyzed in patient serum at baseline (prior to study PN) and over 28 days of follow up. RESULTS eHSP70 declined over time in survivors during 28 days follow-up, but non-survivors had significantly higher eHSP70 concentrations compared to survivors. In patients developing pneumonia, eHSP70, eHSP27, IL-8, and IL-6 were significantly elevated. Adjusted relative risk for hospital mortality was reduced 75% (RR = 0.25, p = 0.001) for SICU patients with a faster decline in eHSP70. The area under the receiver operating characteristic curve was 0.85 (95% CI: 0.76 to 0.94) for the final model suggesting excellent discrimination between SICU survivors and non-survivors. GLN-PN did not alter eHSP70 or eHSP27 serum concentrations over time compared to STD-PN. CONCLUSION Our results suggest that serum HSP70 concentration may be an important marker for severity of illness and likelihood of recovery in the SICU. GLN-supplemented-PN did not increase eHSP70.
Collapse
Affiliation(s)
- Paul E Wischmeyer
- Duke University Hospital, Department of Anesthesiology and Duke Clinical Research Institute, 2301 Erwin Rd, Durham, NC 27710, USA.
| | - Rachael A Mintz-Cole
- Duke University Hospital, Department of Anesthesiology and Duke Clinical Research Institute, 2301 Erwin Rd, Durham, NC 27710, USA.
| | - Christine H Baird
- University of Colorado Denver Anschutz Medical Campus, Anesthesiology, 12700 E. 19th Avenue Box 8602, Aurora, CO 80045, USA.
| | - Kirk A Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
| | - Addison K May
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37240, USA.
| | - Harry C Sax
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Kenneth A Kudsk
- Department of Surgery, University of Wisconsin, Schools of Medicine and Public Health, Madison, Madison, WI 53792, USA.
| | - Li Hao
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Phong H Tran
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Clinical and Molecular Nutrition, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Henry M Blumberg
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
| | - Thomas R Ziegler
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Clinical and Molecular Nutrition, Emory University School of Medicine, Atlanta, GA 30322, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA.
| |
Collapse
|
20
|
Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Protective role of heat shock transcription factor 1 in heart failure: A diagnostic approach. J Cell Physiol 2018; 234:7764-7770. [DOI: 10.1002/jcp.27639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/02/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology Faculty of Medicine, Semnan University of Medical Sciences Semnan Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Reza Shirzad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| |
Collapse
|
21
|
Ranek MJ, Stachowski MJ, Kirk JA, Willis MS. The role of heat shock proteins and co-chaperones in heart failure. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0530. [PMID: 29203715 DOI: 10.1098/rstb.2016.0530] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2017] [Indexed: 12/18/2022] Open
Abstract
The ongoing contractile and metabolic demands of the heart require a tight control over protein quality control, including the maintenance of protein folding, turnover and synthesis. In heart disease, increases in mechanical and oxidative stresses, post-translational modifications (e.g., phosphorylation), for example, decrease protein stability to favour misfolding in myocardial infarction, heart failure or ageing. These misfolded proteins are toxic to cardiomyocytes, directly contributing to the common accumulation found in human heart failure. One of the critical class of proteins involved in protecting the heart against these threats are molecular chaperones, including the heat shock protein70 (HSP70), HSP90 and co-chaperones CHIP (carboxy terminus of Hsp70-interacting protein, encoded by the Stub1 gene) and BAG-3 (BCL2-associated athanogene 3). Here, we review their emerging roles in the maintenance of cardiomyocytes in human and experimental models of heart failure, including their roles in facilitating the removal of misfolded and degraded proteins, inhibiting apoptosis and maintaining the structural integrity of the sarcomere and regulation of nuclear receptors. Furthermore, we discuss emerging evidence of increased expression of extracellular HSP70, HSP90 and BAG-3 in heart failure, with complementary independent roles from intracellular functions with important therapeutic and diagnostic considerations. While our understanding of these major HSPs in heart failure is incomplete, there is a clear potential role for therapeutic modulation of HSPs in heart failure with important contextual considerations to counteract the imbalance of protein damage and endogenous protein quality control systems.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - Marisa J Stachowski
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Chicago, IL 60302, USA
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Chicago, IL 60302, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, CB#7525, Chapel Hill, NC 27599-7525, USA
| |
Collapse
|
22
|
Edkins AL, Price JT, Pockley AG, Blatch GL. Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0521. [PMID: 29203706 DOI: 10.1098/rstb.2016.0521] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
Many heat shock proteins (HSPs) are essential to survival as a consequence of their role as molecular chaperones, and play a critical role in maintaining cellular proteostasis by integrating the fundamental processes of protein folding and degradation. HSPs are arguably among the most prominent classes of proteins that have been broadly linked to many human disorders, with changes in their expression profile and/or intracellular/extracellular location now being described as contributing to the pathogenesis of a number of different diseases. Although the concept was initially controversial, it is now widely accepted that HSPs have additional biological functions over and above their role in proteostasis (so-called 'protein moonlighting'). Most importantly, these new insights are enlightening our understanding of biological processes in health and disease, and revealing novel and exciting therapeutic opportunities. This theme issue draws on therapeutic insights from established research on HSPs in cancer and other non-communicable disorders, with an emphasis on how the intracellular function of HSPs contrasts with their extracellular properties and function, and interrogates their potential diagnostic and therapeutic value to the prevention, management and treatment of chronic diseases.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Adrienne L Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - John T Price
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, University of Melbourne and Western Health, Melbourne, Victoria, Australia.,Department of Medicine, Melbourne Medical School-Western Precinct, University of Melbourne, St Albans, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - A Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Clifton Campus, Clifton Lane, Nottingham, UK
| | - Gregory L Blatch
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa .,Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia.,The Vice Chancellery, The University of Notre Dame Australia, Fremantle, Western Australia, Australia
| |
Collapse
|
23
|
Abstract
T-cell infiltration and the subsequent increased intracardial chronic inflammation play crucial roles in the development of cardiac hypertrophy and heart failure (HF). A77 1726, the active metabolite of leflunomide, has been reported to have powerful anti-inflammatory and T cell-inhibiting properties. However, the effect of A77 1726 on cardiac hypertrophy remains completely unknown. Herein, we found that A77 1726 treatment attenuated pressure overload or angiotensin II (Ang II)-induced cardiac hypertrophy in vivo, as well as agonist-induced hypertrophic response of cardiomyocytes in vitro In addition, we showed that A77 1726 administration prevented induction of cardiac fibrosis by inhibiting cardiac fibroblast (CF) transformation into myofibroblast. Surprisingly, we found that the protective effect of A77 1726 was not dependent on its T lymphocyte-inhibiting property. A77 1726 suppressed the activation of protein kinase B (AKT) signaling pathway, and overexpression of constitutively active AKT completely abolished A77 1726-mediated cardioprotective effects in vivo and in vitro Pretreatment with siRNA targetting Fyn (si Fyn) blunted the protective effect elicited by A77 1726 in vitro More importantly, A77 1726 was capable of blocking pre-established cardiac hypertrophy in mice. In conclusion, A77 1726 attenuated cardiac hypertrophy and cardiac fibrosis via inhibiting FYN/AKT signaling pathway.
Collapse
|
24
|
Exercise Training under Exposure to Low Levels of Fine Particulate Matter: Effects on Heart Oxidative Stress and Extra-to-Intracellular HSP70 Ratio. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9067875. [PMID: 29387296 PMCID: PMC5745714 DOI: 10.1155/2017/9067875] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/19/2017] [Indexed: 01/09/2023]
Abstract
Fine particulate matter (PM2.5) promotes heart oxidative stress (OS) and evokes anti-inflammatory responses observed by increased intracellular 70 kDa heat shock proteins (iHSP70). Furthermore, PM2.5 increases the levels of these proteins in extracellular fluids (eHSP70), which have proinflammatory roles. We investigated whether moderate and high intensity training under exposure to low levels of PM2.5 modifies heart OS and the eHSP70 to iHSP70 ratio (H-index), a biomarker of inflammatory status. Male mice (n = 32), 30 days old, were divided into six groups for 12 weeks: control (CON), moderate (MIT) and high intensity training (HIT), exposure to 5 μg of PM2.5 daily (PM2.5), and moderate and high intensity training exposed to PM2.5 (MIT + PM2.5 and HIT + PM2.5 groups). The CON and PM2.5 groups remained sedentary. The MIT + PM2.5 group showed higher heart lipid peroxidation levels than the MIT and PM2.5 groups. HIT and HIT + PM2.5 showed higher heart lipid peroxidation levels and lower eHSP70 and H-index levels compared to sedentary animals. No alterations were found in heart antioxidant enzyme activity or iHSP70 levels. Moderate exercise training under exposure to low levels of PM2.5 induces heart OS but does not modify eHSP70 to iHSP70 ratio (H-index). High intensity exercise training promotes anti-inflammatory profile despite exposure to low levels of PM2.5.
Collapse
|
25
|
Mortalin deficiency suppresses fibrosis and induces apoptosis in keloid spheroids. Sci Rep 2017; 7:12957. [PMID: 29021584 PMCID: PMC5636810 DOI: 10.1038/s41598-017-13485-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/26/2017] [Indexed: 12/02/2022] Open
Abstract
Mortalin (Mot) is a mitochondrial chaperone of the heat shock protein 70 family and it’s pro-proliferative and anti-apoptosis functions could be associated with keloid pathogenesis, and blocking of mortalin and its interaction with p53 might be a potential novel target for the treatment of keloid. Therefore, we generated mortalin-specific small hairpin (sh) RNAs (dE1-RGD/GFP/shMot) and introduced into keloid spheroids for examination of its apoptotic and anti-fibrotic effect. On keloid tissues, mortalin expression was higher than adjacent normal tissues and it’s protein expressions were activated keloid fibroblasts (KFs). After primary keloid spheroid were transduced with dE1-RGD/GFP/shMot for knockdown of mortalin, expression of type I, III collagen, fibronectin, and elastin was significantly reduced and transforming growth factor-β1, epidermal growth factor receptor (EGFR), Extracellular Signal-Regulated Kinases 1 and 2 (Erk 1/2), and Smad 2/3 complex protein expression were decreased. In addition, increased TUNEL activities and cytochrome C were observed. Further, for examine of mortalin and p53 interaction, we performed immunofluorescence analysis. Knockdown of mortalin relocated p53 to the cell nucleus in primary keloid spheroids by dE1-RGD/GFP/shMot transduction. These results support the utility of knockdown of mortalin to induce apoptosis and reduce ECMs expression in keloid spheroid, which may be highly beneficial in treating keloids.
Collapse
|
26
|
Ehrentraut H, Felix Ehrentraut S, Boehm O, El Aissati S, Foltz F, Goelz L, Goertz D, Kebir S, Weisheit C, Wolf M, Meyer R, Baumgarten G. Tlr4 Deficiency Protects against Cardiac Pressure Overload Induced Hyperinflammation. PLoS One 2015; 10:e0142921. [PMID: 26588247 PMCID: PMC4654478 DOI: 10.1371/journal.pone.0142921] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/28/2015] [Indexed: 12/26/2022] Open
Abstract
Transverse aortic constriction provokes a pro-inflammatory reaction and results in cardiac hypertrophy. Endogenous ligands contribute to cardiac hypertrophy via toll-like receptor (TLR)-4 binding. A lack of TLR4 signaling diminishes hypertrophy and inflammation. Wild type mice undergoing aortic constriction respond to a lipopolysaccharide second-hit stimulus with hyperinflammation. The objective of this study was to assess whether other second-hit challenges utilizing TLR ligands provoke a comparable inflammatory reaction, and to find out whether this response is absent in TLR4 deficient mice. Assuming that cardiac stress alters the expression of pattern recognition receptors we analyzed the effects of transverse aortic constriction and second-hit virulence factor treatment on TLR expression, as well as cytokine regulation. Wild type and Tlr4-/- mice were subjected to three days of TAC and subsequently confronted with gram-positive TLR2 ligand lipoteichoic acid (LTA, 15 mg/g bodyweight) or synthetic CpG-oligodesoxynucleotide 1668 thioate (20 nmol/kg bodyweight, 30 min after D-galactosamin desensitization) signaling via TLR9. Hemodynamic measurements and organ preservation were performed 6 h after stimulation. Indeed, the study revealed a robust enhancement of LTA induced pattern recognition receptor and cytokine mRNA expression and a LTA-dependent reduction of hemodynamic pressure in TAC wild type mice. Second-Hit treatment with CpG-ODNs led to similar results. However, second-hit effects were abolished in Tlr4-/- mice. In total, these data indicate for the first time that cardiac stress increases the inflammatory response towards both, gram-negative and gram-positive, TLR ligands as well as bacterial DNA. The decrease of the inflammatory response upon TLR2 and -9 ligand challenge in TAC Tlr4-/- mice demonstrates that a lack of TLR4 signaling does not only prevent left ventricular hypertrophy but also protects the mice from a cardiac stress induced hyperinflammatory reaction.
Collapse
Affiliation(s)
- Heidi Ehrentraut
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| | - Stefan Felix Ehrentraut
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| | - Olaf Boehm
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| | - Sakina El Aissati
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
- Marienhaus Klinikum, Bad Neuenahr-Ahrweiler, Germany
| | - Fabian Foltz
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
- Asklepios Klinik St. Augustin, Sankt Augustin, Germany
| | - Lina Goelz
- Polyclinic of Orthodontics, University of Bonn, Welschnonnenstraße 17, 53111, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115, Bonn, Germany
| | - David Goertz
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
- Klinik für Orthopädie und Unfallchirurgie, HELIOS Medical Center Siegburg, Siegburg, Germany
| | - Sied Kebir
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Christina Weisheit
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| | - Michael Wolf
- Polyclinic of Orthodontics, University of Bonn, Welschnonnenstraße 17, 53111, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Rainer Meyer
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Georg Baumgarten
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| |
Collapse
|
27
|
Marino LV, Pathan N, Meyer RW, Wright VJ, Habibi P. An in vitro model to consider the effect of 2 mM glutamine and KNK437 on endotoxin-stimulated release of heat shock protein 70 and inflammatory mediators. Nutrition 2015; 32:375-83. [PMID: 26706024 DOI: 10.1016/j.nut.2015.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/13/2015] [Accepted: 09/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Glutamine has been shown to promote the release of heat shock protein 70 (HSP70) both within experimental in vitro models of sepsis and in adults with septic shock. This study aimed to investigate the effects of 2 mM glutamine and an inhibitor of HSP70 (KNK437) on the release of HSP70 and inflammatory mediators in healthy adult volunteers. METHODS An in vitro whole blood endotoxin stimulation assay was used. RESULTS The addition of 2 mM glutamine significantly increased HSP70 levels over time (P < 0.05). HSP70 release had a positive correlation at 4 h with IL-1 β (r = 0.51, P = 0.03) and an inverse correlation with TNF-α (r = -0.56, P = 0.02) and IL-8 levels (r = -0.52, P = 0.03), and there were no significant correlations between HSP70 and IL6 or IL-10 or glutamine. Glutamine supplementation significantly (P < 0.05) attenuated the release of IL-10 at 4 h and IL-8 at 24 h, compared with conditions without glutamine. In endotoxin-stimulated blood there were no significant differences in the release of IL-6, TNF-α, and IL-1 β with glutamine supplementation at 4 and 24 h. However, glutamine supplementation (2 mM) appeared to attenuate the release of inflammatory mediators (IL-1 β, IL-6, TNF-α), although this effect was not statistically significant. The addition of KNK437, a HSP70 inhibitor, significantly diminished HSP70 release, which resulted in lower levels of inflammatory mediators (P < 0.05). CONCLUSION Glutamine supplementation promotes HSP70 release in an experimental model of sepsis. After the addition of KNK437, the effects of glutamine on HSP70 and inflammatory mediator release appear to be lost, suggesting that HSP70 in part orchestrates the inflammatory mediator response to sepsis. The clinical implications require further investigation.
Collapse
Affiliation(s)
- Luise V Marino
- Department of Paediatrics, Imperial College London, London, United Kingdom.
| | - Nazima Pathan
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, Cambridge, United Kingdom
| | - Rosan W Meyer
- Department of Gastroenterology, Great Ormond Street Hospital for Sick Children, London, United Kingdom
| | - Victoria J Wright
- Department of Paediatrics, Imperial College London, London, United Kingdom
| | - Parviz Habibi
- Department of Paediatrics, Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
Jheng HF, Tsai PJ, Chuang YL, Shen YT, Tai TA, Chen WC, Chou CK, Ho LC, Tang MJ, Lai KTA, Sung JM, Tsai YS. Albumin stimulates renal tubular inflammation through an HSP70-TLR4 axis in mice with early diabetic nephropathy. Dis Model Mech 2015; 8:1311-21. [PMID: 26398934 PMCID: PMC4610229 DOI: 10.1242/dmm.019398] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/29/2015] [Indexed: 02/06/2023] Open
Abstract
Increased urinary albumin excretion is not simply an aftermath of glomerular injury, but is also involved in the progression of diabetic nephropathy (DN). Whereas Toll-like receptors (TLRs) are incriminated in the renal inflammation of DN, whether and how albumin is involved in the TLR-related renal inflammatory response remains to be clarified. Here, we showed that both TLR2 and TLR4, one of their putative endogenous ligands [heat shock protein 70 (HSP70)] and nuclear factor-κB promoter activity were markedly elevated in the kidneys of diabetic mice. A deficiency of TLR4 but not of TLR2 alleviated albuminuria, tubulointerstitial fibrosis and inflammation induced by diabetes. The protection against renal injury in diabetic Tlr4−/− mice was associated with reduced tubular injuries and preserved cubilin levels, rather than amelioration of glomerular lesions. In vitro studies revealed that albumin, a stronger inducer than high glucose (HG), induced the release of HSP70 from proximal tubular cells. HSP70 blockade ameliorated albumin-induced inflammatory mediators. HSP70 triggered the production of inflammatory mediators in a TLR4-dependent manner. Moreover, HSP70 inhibition in vivo ameliorated diabetes-induced albuminuria, inflammatory response and tubular injury. Finally, we found that individuals with DN had higher levels of TLR4 and HSP70 in the dilated tubules than non-diabetic controls. Thus, activation of the HSP70-TLR4 axis, stimulated at least in part by albumin, in the tubular cell is a newly identified mechanism associated with induction of tubulointerstitial inflammation and aggravation of pre-existing microalbuminuria in the progression of DN. Summary: Activation of the HSP70-TLR4 axis by albumin in the tubular cell induces tubular inflammation and injury.
Collapse
Affiliation(s)
- Huei-Fen Jheng
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan Institute of Clinical Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Yi-Lun Chuang
- Department of Physiology, National Cheng Kung University, Tainan 701, Taiwan
| | - Yi-Ting Shen
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Ting-An Tai
- Department of Physiology, National Cheng Kung University, Tainan 701, Taiwan
| | - Wen-Chung Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Chuan-Kai Chou
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei 115, Taiwan
| | - Li-Chun Ho
- Division of Nephrology, Department of Internal Medicine, E-DA Hospital/I-Shou University, Kaohsiung 824, Taiwan
| | - Ming-Jer Tang
- Department of Physiology, National Cheng Kung University, Tainan 701, Taiwan
| | | | - Junne-Ming Sung
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Yau-Sheng Tsai
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan Institute of Clinical Medicine, National Cheng Kung University, Tainan 701, Taiwan Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan, Republic of China
| |
Collapse
|
29
|
Bai X, Qi Z, Song G, Zhao X, Zhao H, Meng X, Liu C, Bing W, Bi Y. Effects of Monocyte Chemotactic Protein-1 and Nuclear Factor of Kappa B Pathway in Rejection of Cardiac Allograft in Rat. Transplant Proc 2015; 47:2010-6. [DOI: 10.1016/j.transproceed.2015.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/26/2015] [Accepted: 05/14/2015] [Indexed: 01/04/2023]
|
30
|
Tsai TN, Lee TY, Liu MS, Chuang IC, Lu MC, Dong HP, Lue SI, Yang RC. Release of endogenous heat shock protein 72 on the survival of sepsis in rats. J Surg Res 2015; 198:165-74. [PMID: 26073348 DOI: 10.1016/j.jss.2015.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/17/2015] [Accepted: 05/01/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND This study was undertaken to clarify the role of extracellular heat shock protein 72 on the survival of sepsis and to determine possible factor(s) that may be responsible for it. MATERIALS AND METHODS Sepsis was induced by cecal ligation and puncture. Changes in serum levels of heat shock protein (Hsp72) and cytokines were determined during sepsis, and the results were correlated with the survival. Effects of heat pretreatment on Hsp72 expression in septic rat leukocytes and those of septic rat serum, lipopolysaccharide (LPS), and certain cytokines on the release of Hsp72 in macrophage NR8383 cells were determined. RESULTS Circulating Hsp72 levels were increased during the progress of sepsis (0, 5.5, 6.5, 10, and 6.5 ng/mL at 0, 3, 6, 9, and 18 h after cecal ligation and puncture, respectively) and the increases were correlated positively with survival rates. LPS triggered the release of Hsp72 in heat pretreated animals. Heat pretreatment increased Hsp72 expression in nonsepsis (+535%, P < 0.01) and sepsis (+116%, P<0.01%) rat leukocytes. Incubation of sepsis rat serum with NR8383 cells increased levels of extracellular heat shock protein 72 in cultured medium. Cytokine profiling revealed that among the 19 cytokines screened, four of them were increased as follows: cytokine-induced neutrophil chemoattractant 3 (+211.3%, P < 0.05), interleukin 10 (+147%, P < 0.05), MCP-1 (+49.6%, P < 0.05), and tumor necrosis factor alpha (+51.8%, P < 0.05). MCP-1 and LPS were capable of releasing Hsp72 from NR8383 cells. CONCLUSIONS These results demonstrate that the increases in the levels of circulating Hsp72 had a beneficial effect in improving animal survival during the progress of sepsis. The increases in circulating Hsp72 may be mediated via MCP-1 and/or LPS.
Collapse
Affiliation(s)
- Tsen-Ni Tsai
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Ying Lee
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Maw-Shung Liu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Chun Chuang
- Department of Respiratory Therapy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Chin Lu
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung, Taiwan
| | - Huei-Ping Dong
- Department of Physical Therapy, Fooyin University, Kaohsiung, Taiwan
| | - Sheng-I Lue
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Rei-Chen Yang
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Pediatrics, Changhua Christian Hospital, Changhua, Taiwan.
| |
Collapse
|
31
|
Crozatier B, Ventura-Clapier R. Inhibition of Hypertrophy, Per Se, May Not Be a Good Therapeutic Strategy in Ventricular Pressure Overload: Other Approaches Could Be More Beneficial. Circulation 2015; 131:1448-57. [DOI: 10.1161/circulationaha.114.013895] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Bertrand Crozatier
- From Université Paris-Sud 11, and Institut National de la Santé et de la Recherche Médicale, Unit 1180, Châtenay-Malabry, France
| | - Renée Ventura-Clapier
- From Université Paris-Sud 11, and Institut National de la Santé et de la Recherche Médicale, Unit 1180, Châtenay-Malabry, France
| |
Collapse
|
32
|
Gao L, Huang K, Jiang DS, Liu X, Huang D, Li H, Zhang XD, Huang K. Novel Role for Caspase-Activated DNase in the Regulation of Pathological Cardiac Hypertrophy. Hypertension 2015; 65:871-81. [PMID: 25646292 DOI: 10.1161/hypertensionaha.114.04806] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Lu Gao
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Kun Huang
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Ding-Sheng Jiang
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Xiaoxiong Liu
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Dan Huang
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Hongliang Li
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Xiao-Dong Zhang
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| | - Kai Huang
- From the Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.G., Kun Huang, D.H., Kai Huang); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); Cardiovascular Research Institute, Wuhan University, Wuhan, China (D.-S.J., X.L., H.L.); and Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China (X.-D.Z.)
| |
Collapse
|
33
|
Liu H, Xie Q, Xin BM, Liu JL, Liu Y, Li YZ, Wang JP. Inhibition of autophagy recovers cardiac dysfunction and atrophy in response to tail-suspension. Life Sci 2014; 121:1-9. [PMID: 25476825 DOI: 10.1016/j.lfs.2014.10.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/30/2014] [Accepted: 10/28/2014] [Indexed: 12/21/2022]
Abstract
AIMS Physical inactivity during space flight or prolonged bed rest may cause cardiac dysfunction and atrophy, but the exact mechanism that governs the regulation of myocardial dysfunction and cardiac atrophy remains poorly understood. Autophagy, a protein degradation pathway, has recently been shown to be involved in the regulation of cardiac dysfunction and atrophy. In this study, we investigated the relationships between dysfunction and inactivity-induced atrophy and autophagy in rat cardiac tissue. MAIN METHODS Physical inactivity was simulated by a tail suspension model, and cardiac function was examined by echocardiography. Cardiac atrophy was measured by wheat germ agglutinin staining and autophagic activity was detected by Western blot analysis and immunofluorescence staining. KEY FINDINGS We demonstrated that cardiac function, especially contractility, declined and the area of cardiac atrophy increased in the tail-suspended cardiac tissue. Additionally, the cross-sectional area of myocardial cells decreased; however, apoptosis did not increase with tail suspension. Similarly, the expression of autophagy-related proteins and the number of autophagosomes were elevated in the tail-suspended cardiac tissue. Moreover, the administration of chloroquine, an autophagy inhibitor, reversed cardiac dysfunction and atrophy via the suppression of autophagic activity during suspension. Our results indicate that autophagy facilitates the development and progression of cardiac dysfunction and atrophy induced by tail suspension. SIGNIFICANCE Our studies hint that the components of the autophagy-related signaling pathway are potential therapeutic targets for the treatment of cardiac diseases induced by physical inactivity.
Collapse
Affiliation(s)
- Hong Liu
- Department of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College, Chinese Academy of Medical Sciences, 1# Tiantan Xili, Beijing 100050, PR China
| | - Qiong Xie
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China
| | - Bing-Mu Xin
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China
| | - Jun-Lian Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China
| | - Yu Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China
| | - Yong-Zhi Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China
| | - Jia-Ping Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Centre, 26# Beiqing Road, Beijing 100094, PR China.
| |
Collapse
|
34
|
Bellaye PS, Burgy O, Causse S, Garrido C, Bonniaud P. Heat shock proteins in fibrosis and wound healing: Good or evil? Pharmacol Ther 2014; 143:119-32. [DOI: 10.1016/j.pharmthera.2014.02.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 12/22/2022]
|
35
|
Chaperone heat shock protein 70 in nucleus accumbens core: a novel biological target of behavioural sensitization to morphine in rats. Int J Neuropsychopharmacol 2014; 17:469-84. [PMID: 24280010 DOI: 10.1017/s1461145713001429] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Drug addiction is a major public health issue, yet the underlying adaptation of neural networks by drugs of abuse is not fully understood. We have previously linked chaperone heat shock protein 70 (Hsp70) to drug-induced adaptations. Focusing on the NAc core and shell, the present study aims to provide further findings for our understanding of the relation between behavioural sensitization to morphine and Hsp70 at transcriptional and functional levels in rats. Firstly, we delineated the characteristics of behavioural sensitization induced by a single morphine exposure (1-10 mg/kg, s.c.). Secondly, Hsp70 protein expression in the NAc core was time- and dose-relatedly induced during the development of behavioural sensitization to a single morphine exposure in rats, and Pearson analysis indicated a positive correlation between behavioural sensitization and Hsp70 expression in NAc core. Thirdly, at the transcriptional level, intra-NAc core injection of the specific heat shock factor-I (HSF-I) inhibitor N-Formyl-3,4-methylenedioxy-benzylidine-γ-butyrolactam (KNK437) suppressed Hsp70 expression and the development of behavioural sensitization, while the HSF-I specific inducer geranylgeranylacetone (GGA) promoted both of them. Interestingly, intra-NAc shell injection of KNK437 or GGA did not affect the development of behavioural sensitization. Finally, both the functional inhibition of Hsp70 ATPase activity by methylene blue (MB), and the antagonism of Hsp70 substrate binding site (SBD) activity by pifithrin-μ (PES) impaired the development of behavioural sensitization when they were microinjected into the NAc core. Taken together, the critical involvement of chaperone Hsp70 in behavioural sensitization to morphine identifies a biological target for long-lasting adaptations with relevance to addiction.
Collapse
|
36
|
Ghigo A, Franco I, Morello F, Hirsch E. Myocyte signalling in leucocyte recruitment to the heart. Cardiovasc Res 2014; 102:270-80. [PMID: 24501328 DOI: 10.1093/cvr/cvu030] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myocardial damage, by different noxious causes, triggers an inflammatory reaction driving post-injury repair mechanisms and chronic remodelling processes that are largely detrimental to cardiac function. Cardiomyocytes have recently emerged as key players in orchestrating this inflammatory response. Injured cardiomyocytes release damage-associated molecular pattern molecules, such as high-mobility group box 1 (HMGB1), DNA fragments, heat shock proteins, and matricellular proteins, which instruct surrounding healthy cadiomyocytes to produce inflammatory mediators. These mediators, mainly interleukin (IL)-1β, IL-6, macrophage chemoattractant protein (MCP)-1, and tumour necrosis factor α (TNF-α), in turn activate versatile signalling networks within surviving cardiomyocytes and trigger leucocyte activation and recruitment. In this review, we will focus on recently characterized signalling pathways activated in cardiomyocytes that mediate inflammatory responses during myocardial infarction, hypertensive heart disease, and myocarditis.
Collapse
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino, Italy
| | | | | | | |
Collapse
|
37
|
Lin L, Knowlton AA. Innate immunity and cardiomyocytes in ischemic heart disease. Life Sci 2014; 100:1-8. [PMID: 24486305 DOI: 10.1016/j.lfs.2014.01.062] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/07/2014] [Accepted: 01/15/2014] [Indexed: 02/07/2023]
Abstract
Myocardial ischemia/reperfusion (I/R) is the most common cause of myocardial inflammation, which is primarily a manifestation of the innate immune responses. Innate immunity is activated when pattern recognition receptors (PRRs) respond to molecular patterns common to microbes and to danger signals expressed by injured or infected cells, so called pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The expression of various PRRs in cardiomyocytes and the release of DAMPs from cardiomyocytes subjected to I/R injury, through active mechanisms as well as passive processes, enable cardiomyocytes to generate innate immune responses. Studies in isolated heart and cardiomyocytes have confirmed the inflammatory and functional effects of cardiac PRRs especially Toll-like receptors in response to I/R-derived DAMPs, such as heat shock proteins. This review addresses the active role of cardiomyocytes in mediating innate inflammatory responses to myocardial I/R. We propose that cardiomyocytes act as innate immune cells in myocardial I/R injury.
Collapse
Affiliation(s)
- Li Lin
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Anne A Knowlton
- Molecular and Cellular Cardiology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; Department of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; Department of Pharmacology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; The Northern California VA, Sacramento, CA, USA.
| |
Collapse
|
38
|
Luzina IG, Kopach P, Lockatell V, Kang PH, Nagarsekar A, Burke AP, Hasday JD, Todd NW, Atamas SP. Interleukin-33 potentiates bleomycin-induced lung injury. Am J Respir Cell Mol Biol 2014; 49:999-1008. [PMID: 23837438 DOI: 10.1165/rcmb.2013-0093oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The mechanisms of interstitial lung disease (ILD) remain incompletely understood, although recent observations have suggested an important contribution by IL-33. Substantial elevations in IL-33 expression were found in the lungs of patients with idiopathic pulmonary fibrosis and scleroderma lung disease, as well as in the bleomycin injury mouse model. Most of the observed IL-33 expression was intracellular and intranuclear, suggesting involvement of the full-length (fl) protein, but not of the proteolytically processed mature IL-33 cytokine. The effects of flIL-33 on mouse lungs were assessed independently and in combination with bleomycin injury, using recombinant adenovirus-mediated gene delivery. Bleomycin-induced changes were not affected by gene deficiency of the IL-33 receptor T1/ST2. Combined flIL-33 expression and bleomycin injury exerted a synergistic effect on pulmonary lymphocyte and collagen accumulation, which could be explained by synergistic regulation of the cytokines transforming growth factor-β, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein\x{2013}1α, and tumor necrosis factor-α. By contrast, no increase in the levels of the Th2 cytokines IL-4, IL-5, or IL-13 was evident. Moreover, flIL-33 was found to increase the expression of several heat shock proteins (HSPs) significantly, and in particular HSP70, which is known to be associated with ILD. Thus, flIL-33 is a synergistic proinflammatory and profibrotic regulator that acts by stimulating the expression of several non-Th2 cytokines, and activates the expression of HSP70.
Collapse
Affiliation(s)
- Irina G Luzina
- 1 Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore; and
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dong J, Guo L, Liao Z, Zhang M, Zhang M, Wang T, Chen L, Xu D, Feng Y, Wen F. Increased expression of heat shock protein 70 in chronic obstructive pulmonary disease. Int Immunopharmacol 2013; 17:885-93. [PMID: 24095952 DOI: 10.1016/j.intimp.2013.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 08/24/2013] [Accepted: 09/04/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Heat shock protein 70 (HSP70) plays a critical role in the process of inflammation and innate immunity response under environmental stress. OBJECTIVES This study was to investigate HSP70 expression in the peripheral lung tissues of chronic obstructive pulmonary disease (COPD) patients and in human bronchial epithelial cells (16-HBE) exposed to cigarette smoke extract (CSE). METHODS Peripheral lung tissues were collected after lung cancer resection from 26 patients without COPD, 20 with mild COPD and 15 with advanced COPD, classified by lung function criteria. Among these cases, 37 were smokers and 24 non-smokers. Lung tissues were examined for histopathological changes and levels of HSP70 and IL-8. Cultured 16-HBE cells were stimulated with CSE in the absence or presence of HSP70 neutralizing antibody and the expressions of IL-8 and phospho-EGFR protein were determined. RESULTS Compared to patients without COPD, the levels of HSP70 and IL-8 were significantly increased in the lung tissues of COPD patients and positively correlated with the severity of the disease. The HSP70 expression was significantly higher in current smokers than that in non-smokers. Moreover, CSE-induced HSP70 significantly enhanced IL-8 production and EGFR phosphorylation in 16-HBE cells. The increases in IL-8 and phospho-EGFR were blocked by anti-HSP70 antibody. CONCLUSIONS Our study clarified that increased expression of HSP70 is closely related to COPD disease severity and smoking status. Extracellular HSP70 regulated chemokine productions and EGFR phosphorylation and plays an important role in the CSE-induced inflammatory and innate immunity responses in bronchial epithelia cells.
Collapse
Affiliation(s)
- Jiajia Dong
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China and Department of Respiratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hassanpour H, Afzali A, Bahadoran S. Diminished gene expression of cardiac heat shock proteins in pulmonary hypertensive chickens. Br Poult Sci 2013; 54:581-6. [DOI: 10.1080/00071668.2013.828151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Yuan Y, Zong J, Zhou H, Bian ZY, Deng W, Dai J, Gan HW, Yang Z, Li H, Tang QZ. Puerarin attenuates pressure overload-induced cardiac hypertrophy. J Cardiol 2013; 63:73-81. [PMID: 23906530 DOI: 10.1016/j.jjcc.2013.06.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/22/2013] [Accepted: 06/09/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND Puerarin is the most abundant isoflavonoid in kudzu root. It has been used to treat angina pectoris and myocardial infarction clinically. However, little is known about the effect of puerarin on cardiac hypertrophy. METHODS Aortic banding (AB) was performed to induce cardiac hypertrophy in mice. Puerarin premixed in diets was administered to mice after one week of AB. Echocardiography and catheter-based measurements of hemodynamic parameters were performed at 7 weeks after starting puerarin treatment (8 weeks post-surgery). The extent of cardiac hypertrophy was also evaluated by pathological and molecular analyses of heart samples. Cardiomyocyte apoptosis was assessed by measuring Bax and Bcl-2 protein expression and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. In addition, the inhibitory effect of puerarin (1 μM, 5 μM, 10 μM, 20 μM, 40 μM) on mRNA expression of atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) in Ang II (1 μM)-stimulated H9c2 cells was investigated using quantitative real-time reverse transcription-polymerase chain reaction. RESULTS Echocardiography and catheter-based measurements of hemodynamic parameters at 7 weeks revealed the amelioration of systolic and diastolic abnormalities. Puerarin also decreased cardiac fibrosis in AB mice. Moreover, the beneficial effect of puerarin was associated with the normalization in gene expression of hypertrophic and fibrotic markers. Further studies showed that pressure overload significantly induced the activation of phosphoinositide 3-kinase (PI3K)/Akt signaling and c-Jun N-terminal kinase (JNK) signaling, which was blocked by puerarin treatment. Cardiomyocyte apoptosis and induction of Bax in response to AB were suppressed by puerarin. Furthermore, the increased mRNA expression of ANP and BNP induced by Ang II (1 μM) was restrained to a different extent by different concentrations of puerarin. CONCLUSION Puerarin may have an ability to retard the progression of cardiac hypertrophy and apoptosis which is probably mediated by the blockade of PI3K/Akt and JNK signaling pathways.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Jing Zong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Zhou-Yan Bian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Jia Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hua-Wen Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Zheng Yang
- 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
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China.
| |
Collapse
|
42
|
Li Z, Song Y, Xing R, Yu H, Zhang Y, Li Z, Gao W. Heat shock protein 70 acts as a potential biomarker for early diagnosis of heart failure. PLoS One 2013; 8:e67964. [PMID: 23874478 PMCID: PMC3706597 DOI: 10.1371/journal.pone.0067964] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/24/2013] [Indexed: 11/18/2022] Open
Abstract
Early identification for heart failure (HF) may be useful for disease modifying treatment in order to reduce heart disease progression or even to reverse it. In our previous studies, we have revealed a group of heat shock proteins (HSPs) which might be related to neonatal rat cardiomyocyte hypertrophy by proteomic approach. Here, we confirm that HSPs, including HSP27 and HSP70, altered in the early stage of cardiac remodeling in vivo animal model. Furthermore, plasma concentrations of those HSPs and their potential screening value were evaluated at different stages in 222 patient subjects. Plasma HSP27, HSP70 and HSP90 were measured using enzyme-linked immunosorbent assay. Results indicate that HSP70 was positively correlated to the severity (progression) of HF (r = 0.456, p<0.001). The area under the rate of change (ROC) curve was 0.601 (p = 0.017) in patients with stage B HF and 0.835 (p<0.001) in those with stage C HF. However, HSP27 and HSP90 did not display significant changes in any stage of HF in this study. Taken together, plasma concentrations of HSP70 elevated with the progression of HF and might act as a potential screening biomarker for early diagnosis of HF.
Collapse
Affiliation(s)
- Zongshi Li
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
| | - Yao Song
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
| | - Rui Xing
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
| | - Haiyi Yu
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
| | - Youyi Zhang
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
| | - Zijian Li
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
- * E-mail: (WG); (ZL)
| | - Wei Gao
- Department of Cardiology, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovasicular Receptors Research Beijing, China
- * E-mail: (WG); (ZL)
| |
Collapse
|
43
|
Gruden G, Barutta F, Pinach S, Lorenzati B, Cavallo-Perin P, Giunti S, Bruno G. Circulating anti-Hsp70 levels in nascent metabolic syndrome: the Casale Monferrato Study. Cell Stress Chaperones 2013; 18:353-7. [PMID: 23212539 PMCID: PMC3631095 DOI: 10.1007/s12192-012-0388-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/20/2012] [Accepted: 11/21/2012] [Indexed: 12/20/2022] Open
Abstract
The metabolic syndrome (MetS) confers an increased risk of both type 2 diabetes and cardiovascular diseases (CVD). Heat shock protein 70 (Hsp70), an intracellular polypeptide, can be exposed on the plasma membrane and/or released into the circulation, eliciting both native and immune responses that may contribute to vascular damage. Our aim was to assess if serum anti-Hsp70 antibody levels were cross-sectionally associated with uncomplicated MetS. A cross-sectional case-control study from the nondiabetic cohort of the Casale Monferrato Study was performed. Subjects with established CVD and/or abnormal renal function were excluded. Case subjects (n = 180) were defined as those fulfilling the criteria for the diagnosis of MetS. Control subjects (n = 136) were completely free of any component of the MetS. Serum anti-Hsp70 levels were measured by immunoenzymatic assay. We found that anti-Hsp70 antibody levels were significantly higher in cases than in control subjects [122.6 (89.5-155.6) vs 107.1 (77.3-152.4) μg/ml, p = 0.04], even after age and sex adjustment. In logistic regression analysis, higher levels of log-anti-Hsp70 conferred greater odds ratio (OR) for MetS, independently of age and sex. There was a statistically significant trend of ORs across quartiles of anti-Hsp70 and values greater than 108.0 μg/ml conferred a 77% increased OR of MetS as compared with values in the lower quartiles. The strength of the association slightly decreased after further adjustment for apolipoprotein B, smoking, and albumin excretion rate. In conclusion, our results show that serum anti-Hsp70 antibody levels are independently associated with nascent MetS.
Collapse
Affiliation(s)
- Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Cardioprotective molecules are enriched in beating cardiomyocytes derived from human embryonic stem cells. Int J Cardiol 2013; 165:341-54. [DOI: 10.1016/j.ijcard.2012.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 07/16/2012] [Accepted: 07/21/2012] [Indexed: 01/11/2023]
|
45
|
Du D, Yan J, Ren J, Lv H, Li Y, Xu S, Wang Y, Ma S, Qu J, Tang W, Hu Z, Yu S. Synthesis, biological evaluation, and molecular modeling of glycyrrhizin derivatives as potent high-mobility group box-1 inhibitors with anti-heart-failure activity in vivo. J Med Chem 2012. [PMID: 23199028 DOI: 10.1021/jm301248y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel glycyrrhizin (GL) derivatives were designed and synthesized by introducing various amine or amino acid residues into the carbohydrate chain and at C-30. Their inhibitory effects on high-mobility group box 1 (HMGB1) were evaluated using a cell-based lipopolysaccharide (LPS) induced tumor necrosis factor α (TNF-α) release study. Compounds 10, 12, 18-20, 23, and 24, which had substituents introduced at C-30, demonstrated moderate HMGB1 inhibition with ED₅₀ values ranging from 337 to 141 μM, which are values comparable to that of the leading GL compound (1) (ED₅₀ = 70 μM). Compounds 23 and 24 emerged as novel and interesting HMGB1 inhibitors. These compounds were able to extend the survival of mice with chronic heart failure (CHF) and acute heart failure (AHF), respectively. In addition, molecular modeling studies were performed to support the biological data.
Collapse
Affiliation(s)
- Dan Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dos Santos LF, Antonio EL, Serra AJ, Venturini G, Montemor J, Okada M, Araújo S, Tucci P, de Paola A, Fenelon G. Thermotolerance does not reduce the size or remodeling of radiofrequency lesions in the rat myocardium. J Interv Card Electrophysiol 2012; 36:5-11; discussion 11. [PMID: 23080332 DOI: 10.1007/s10840-012-9746-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 09/14/2012] [Indexed: 11/30/2022]
Abstract
PURPOSE Late lesion extension may be involved in the genesis of delayed radiofrequency (RF) effects. Because RF lesion is thermally mediated, we hypothesized that induction of heat shock response (thermotolerance) would modulate lesion healing. We evaluated the effects of thermotolerance on the dimensions and remodeling of RF lesions in a rat model of heart failure. METHODS Wistar rats (weight 300 g) subjected to heat stress (n = 22, internal temperature of 42 °C for 10 min) were compared to controls (n = 22, internal temperature of 37 °C for 10 min). After 48 h (peak of HSP70 myocardial concentration), a modified unipolar RF lesion (customized catheter, tip 4.5 mm in diameter; 12 W; 10 s) was created on the left ventricular free wall. Animals were sacrificed 2 h (n = 10 per group) and 4 weeks (n = 12 per group) after ablation for lesion analysis. An echocardiogram was obtained at 4 weeks. RESULTS There was no difference between groups regarding the size of acute (controls 27 ± 2 vs. treated 27 ± 3 mm(2)) and chronic lesions (controls 17 ± 1 vs. treated 19 ± 1 mm(2)). Histology of lesions did not differ between groups. The echocardiogram revealed dilation of the cavities and moderate systolic dysfunction without difference between groups. Acute lesion dimensions were similar between control and treated animals over time (ablation undertaken 3, 12, 24, 48, and 72 h after hyperthermia) and also using a conventional ablation catheter (50 °C; 15 W; 10 s). CONCLUSION Thermotolerance does not reduce the size or remodeling of RF lesions in the rat myocardium.
Collapse
Affiliation(s)
- Luís Felipe Dos Santos
- Discipline of Cardiology, Paulista School of Medicine, Federal University of São Paulo, Pedro de Toledo 781, 10th Floor (Cardiology), São Paulo, SP 04039-032, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
HSP70 increases extracellular matrix production by human vascular smooth muscle through TGF-β1 up-regulation. Int J Biochem Cell Biol 2012; 45:232-42. [PMID: 23084979 DOI: 10.1016/j.biocel.2012.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 09/04/2012] [Accepted: 10/09/2012] [Indexed: 01/01/2023]
Abstract
The circulating levels of heat shock proteins (HSP) are increased in cardiovascular diseases; however, the implication of this for the fibrotic process typical of such diseases remains unclear. HSP70 can interact with the vascular smooth muscle cells (SMC), the major producer of extracellular matrix (ECM) proteins, through the Toll-like receptors 4 (TLR4). The transforming growth factor type-β1 (TGF-β1) is a well known vascular pro-fibrotic cytokine that is regulated in part by AP-1-dependent transcriptional mechanisms. We hypothesized that extracellular HSP70 could interact with SMCs, inducing TGF-β1 synthesis and subsequent changes in the vascular ECM. We demonstrate that extracellular HSP70 binds to human aorta SMC TLR4, which up-regulates the AP-1-dependent transcriptional activity of the TGF-β1 promoter. This is achieved through the mitogen activated protein kinases JNK and ERK, as demonstrated by the use of specific blockers and the knockdown of TLR4 with specific small interfering RNAs. The TGF-β1 upregulation increase the expression of the ECM proteins type I collagen and fibronectin. This novel observation may elucidate the mechanisms by which HSP70 contributes in the inflammation and fibrosis present in atherosclerosis and other fibrosis-related diseases.
Collapse
|
48
|
Dulin E, García-Barreno P, Guisasola MC. Genetic variations of HSPA1A, the heat shock protein levels, and risk of atherosclerosis. Cell Stress Chaperones 2012; 17:507-16. [PMID: 22328194 PMCID: PMC3368027 DOI: 10.1007/s12192-012-0328-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/20/2012] [Accepted: 01/25/2012] [Indexed: 10/14/2022] Open
Abstract
HSPA1A is a serum and intracellular heat shock protein with antiapoptotic and antithrombotic properties. The present study examines the hypothesis that a decrease in the synthesis of this protein in relation to certain polymorphisms of the regulatory region of the HSPA1A gene can define a vascular disease risk phenotype. A randomly selected population was studied and stratified into groups according to the degree of vascular risk. After applying the Task Force Chart to 452 people, the subjects were divided into three groups: group 0 (no vascular risk factor or risk < 5%), n = 239; group 1 (moderate (10-20%) risk, with no clinical cardiovascular disease), n = 161; and group 2 (overt atherosclerosis), n = 52. Serum and intragranulocytic HSPA1A was quantified, and direct Sanger sequencing was performed in all subjects. An analysis was made of the association of two single nucleotide polymorphisms (db rs1008438 -110A/C and db rs1043618 +190 G/C) with circulating and intragranulocytic HSPA1A and the risk of atherosclerosis. The atherosclerotic subjects showed significantly lower circulating HSPA1A levels than the other groups, regardless of the genotype. The patients with CC genotype for both polymorphisms showed significantly lower intragranulocytic HSPA1A levels than the other genotypes. Serum HSPA1A concentrations could be proposed as a biomarker of cardiovascular disease. CC homozygosis for polymorphisms db rs1008438 and db rs1043618 is associated with a decrease in the intragranulocytic production of HSPA1A. Given the antiatherogenic functions of intracellular HSPA1A, the -110A and +190 G alleles could constitute potential genetic biomarkers of a less severe clinical phenotype for the risk of developing atherosclerosis.
Collapse
Affiliation(s)
- Elena Dulin
- Biochemistry Department, Hospital General Universitario “Gregorio Marañón”, Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Pedro García-Barreno
- Experimental Medical & Surgery Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario “Gregorio Marañón”, Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Maria C. Guisasola
- Experimental Medical & Surgery Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario “Gregorio Marañón”, Dr. Esquerdo 46, 28007 Madrid, Spain
| |
Collapse
|
49
|
Bharti S, Singh R, Kumar R, Malik S, Hussain T, Al-Attas OS, Arya DS. WITHDRAWN: Hsp70 overexpression coordinately regulates myocardial hypertrophy, fibrosis and contractile function in 5-HT(2B) blockade mediated anti-hypertrophic effect. BIOCHIMICA ET BIOPHYSICA ACTA 2012:S0925-4439(12)00119-6. [PMID: 22659376 DOI: 10.1016/j.bbadis.2012.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 05/10/2012] [Accepted: 05/16/2012] [Indexed: 11/19/2022]
Abstract
This article has been withdrawn at the request of the editors. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Saurabh Bharti
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | | | | | | | | | | | | |
Collapse
|
50
|
Xiao J, Jiang H, Zhang R, Fan G, Zhang Y, Jiang D, Li H. Augmented cardiac hypertrophy in response to pressure overload in mice lacking ELTD1. PLoS One 2012; 7:e35779. [PMID: 22606234 PMCID: PMC3350503 DOI: 10.1371/journal.pone.0035779] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/21/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Epidermal growth factor (EGF), latrophilin and seven transmembrane domain-containing protein 1 (ELTD1) is developmentally upregulated in the heart. Little is known about the relationship between ELTD1 and cardiac diseases. Therefore, we aimed to clarify the role of ELTD1 in pressure overload-induced cardiac hypertrophy. METHODS AND RESULTS C57BL/6J wild-type (WT) mice and ELTD1-knockout (KO) mice were subjected to left ventricular pressure overload by descending aortic banding (AB). KO mice exhibited more unfavorable cardiac remodeling than WT mice 28 days post AB; this remodeling was characterized by aggravated cardiomyocyte hypertrophy, thickening of the ventricular walls, dilated chambers, increased fibrosis, and blunted systolic and diastolic cardiac function. Analysis of signaling pathways revealed enhanced extracellular signal-regulated kinase (ERK) and the c-Jun amino-terminal kinase (JNK) phosphorylation in response to ELTD1 deletion. CONCLUSIONS ELTD1 deficiency exacerbates cardiac hypertrophy and cardiac function induced by AB-induced pressure overload by promoting both cardiomyocyte hypertrophy and cardiac fibrosis. These effects are suggested to originate from the activation of the ERK and JNK pathways, suggesting that ELTD1 is a potential target for therapies that prevent the development of cardiac disease.
Collapse
Affiliation(s)
- Jinfeng Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
- * E-mail: (HJ); (HL)
| | - Rui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
| | - Guangpu Fan
- Department of Thoracic and Cardiac Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
| | - Dingsheng Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People’s Republic of China
- * E-mail: (HJ); (HL)
| |
Collapse
|