1
|
Chen W, Liu L, Tang M, Li J, Yuan W, Yin D, Cao Y, Tian J. Type I collagen-targeted liposome delivery of Serca2a modulates myocardium calcium homeostasis and reduces cardiac fibrosis induced by myocardial infarction. Mater Today Bio 2024; 28:101162. [PMID: 39175654 PMCID: PMC11339061 DOI: 10.1016/j.mtbio.2024.101162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/24/2024] Open
Abstract
Fibrotic scarring and impaired myocardial calcium homeostasis serve as the two main factors in the pathology of heart failure following myocardial infarction (MI), leading to poor prognosis and death in patients. Serca2a is a target of interest in gene therapy for MI-induced heart failure via the regulation of intracellular calcium homeostasis and, subsequently, enhancing myocardial contractility. A recent study also reported that Serca2a ameliorates pulmonary fibrosis by blocking nuclear factor kB (NF-kB)/interleukin-6 (IL-6)-induced (SMAD)/TGF-β signaling activation, while the effect in MI-induced myocardial fibrosis remains to be addressed. Here, we loaded Serca2a plasmids into type 1 collagen-targeting nanoparticles to synthesize the GKWHCTTKFPHHYCLY-Serca2a-Liposome (GSL-NPs) for targeted treatment of myocardial infarction. We showed that GSL-NPs were effectively targeted in the scar area in MI-induced mice within tail-vein delivery for 48 h. Treatment with GSL-NPs improved cardiac functions and shrank fibrotic scars after MI in mice by up-regulating Serca2a. In cardiac fibroblasts, GSL-NPs alleviated hypoxia-induced fibrotic progression partly by inhibiting NF-kB activation. Furthermore, treatment with GSL-NPs protected cardiomyocyte calcium homeostasis and enhanced myocardial contractility during hypoxia. Together, we demonstrate that type I collagen-targeted liposome delivery of Serca2a may benefit patients with myocardial infarction by inhibiting fibrotic scarring as well as modulation of calcium homeostasis.
Collapse
Affiliation(s)
- Wanshi Chen
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Ultrasound Department of Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lingjuan Liu
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajin Li
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenjing Yuan
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Yin
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Ultrasound Department of Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Ultrasound Department of Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jie Tian
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Paediatrics, National Clinical Key Cardiovascular Specialty, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Ni Y, Hua Y, He Z, Hu W, Chen Z, Wang D, Li X, Sun Y, Jiang G. Release of exosomes from injectable silk fibroin and alginate composite hydrogel for treatment of myocardial infarction. J Biomater Appl 2024; 39:139-149. [PMID: 38688330 DOI: 10.1177/08853282241251610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Myocardial infarction (MI) is considered as a significant cause of death globally. Exosomes (EXOs) are essential for intercellular communication and pathophysiology of several cardiovascular diseases. Nevertheless, the short half-life and rapid clearance of EXOs leads to a lack of therapeutic doses delivered to the lesioned area. Therefore, an injectable silk fibroin and alginate (SF/Alg) composite hydrogel was developed to bind folate receptor-targeted EXOs (FA-EXOs) derived from H9C2 cells for the therapy of myocardial injury following myocardial infarction-ischemia/reperfusion (MI-I/R). The resulting composite exhibits a variety of properties, including adjustable gelation kinetics, shear-thinning injectability, soft and dynamic stability that adapts to the heartbeat, and outstanding cytocompatibility. After injected into the damaged rat heart, administration of SF/Alg + FA-EXOs significantly enhanced cardiac function as demonstrated by improved ejection fraction, fractional shortening and decreased fibrosis area. The results of real-time PCR and immunofluorescence staining show a remarkable up-regulation in the expression of proteins (CD31) and genes (VWF and Serca2a) related to the heart. Conversely, expression of fibrosis-related genes (TGF-β1) decreased significantly. Therefore, the obtained SF/Alg + FA-EXOs system remarkably enhanced the intercellular interactions, promoted cell proliferation and angiogenesis, and achieved an outstanding therapeutic effect on MI.
Collapse
Affiliation(s)
- Yunjie Ni
- Department of Cardiology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Yinjian Hua
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, China
| | - Zhengfei He
- Department of Cardiology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Weilv Hu
- Department of Cardiology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Zhiyun Chen
- Department of Cardiology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Diqing Wang
- Department of Cardiology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Xintong Li
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Yanfang Sun
- School of Life Science and Medicines, Zhejiang Sci-Tech University, Hangzhou, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, China
| |
Collapse
|
3
|
Fan W, Sun X, Yang C, Wan J, Luo H, Liao B. Pacemaker activity and ion channels in the sinoatrial node cells: MicroRNAs and arrhythmia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:151-167. [PMID: 36450332 DOI: 10.1016/j.pbiomolbio.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/13/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The primary pacemaking activity of the heart is determined by a spontaneous action potential (AP) within sinoatrial node (SAN) cells. This unique AP generation relies on two mechanisms: membrane clocks and calcium clocks. Nonhomologous arrhythmias are caused by several functional and structural changes in the myocardium. MicroRNAs (miRNAs) are essential regulators of gene expression in cardiomyocytes. These miRNAs play a vital role in regulating the stability of cardiac conduction and in the remodeling process that leads to arrhythmias. Although it remains unclear how miRNAs regulate the expression and function of ion channels in the heart, these regulatory mechanisms may support the development of emerging therapies. This study discusses the spread and generation of AP in the SAN as well as the regulation of miRNAs and individual ion channels. Arrhythmogenicity studies on ion channels will provide a research basis for miRNA modulation as a new therapeutic target.
Collapse
Affiliation(s)
- Wei Fan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Xuemei Sun
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Chao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Hongli Luo
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Bin Liao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
4
|
Hula N, Spaans F, Vu J, Quon A, Kirschenman R, Cooke CLM, Phillips TJ, Case CP, Davidge ST. Placental treatment improves cardiac tolerance to ischemia/reperfusion insult in adult male and female offspring exposed to prenatal hypoxia. Pharmacol Res 2021; 165:105461. [PMID: 33513355 DOI: 10.1016/j.phrs.2021.105461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/08/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Offspring born from complicated pregnancies are at greater risk of cardiovascular disease in adulthood. Prenatal hypoxia is a common pregnancy complication that results in placental oxidative stress and impairs fetal development. Adult offspring exposed to hypoxia during fetal life are more susceptible to develop cardiac dysfunction, and show decreased cardiac tolerance to an ischemia/reperfusion (I/R) insult. To improve offspring cardiac outcomes, we have assessed the use of a placenta-targeted intervention during hypoxic pregnancies, by encapsulating the mitochondrial antioxidant MitoQ into nanoparticles (nMitoQ). We hypothesized that maternal nMitoQ treatment during hypoxic pregnancies improves cardiac tolerance to I/R insult in adult male and female offspring. Pregnant Sprague-Dawley rats were exposed to normoxia (21 % O2) or hypoxia (11 % O2) from gestational day 15-20, after injection with 100 μL saline or nMitoQ (125 μM) on GD15 (n=6-8/group). Male and female offspring were aged to 4 months. Both male and female offspring from hypoxic pregnancies showed reduced cardiac tolerance to I/R (assessed ex vivo using the isolated working heart technique) which was ameliorated by nMitoQ treatment. To identify potential molecular mechanisms for the changes in cardiac tolerance to I/R, cardiac levels/phosphorylation of proteins important for intracellular Ca2+ cycling were assessed with Western blotting. In prenatally hypoxic male offspring, improved cardiac recovery from I/R by nMitoQ was accompanied by increased cardiac phospholamban and phosphatase 2Ce levels, and a trend to decreased Ca2+/calmodulin-dependent protein kinase IIδ phosphorylation. In contrast, in female offspring, nMitoQ treatment in hypoxic pregnancies increased phospholamban and protein kinase Cε phosphorylation. Maternal nMitoQ treatment improves cardiac tolerance to I/R insult in adult offspring and thus has the potential to improve the later-life trajectory of cardiovascular health of adult offspring born from pregnancies complicated by prenatal hypoxia.
Collapse
Affiliation(s)
- Nataliia Hula
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Jennie Vu
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| | - Tom J Phillips
- Dementia Research Institute, Cardiff University, Cardiff, CF10 3AT, UK.
| | - C Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol, BS10 5NB, UK.
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, T6G 2S2, Alberta, Canada.
| |
Collapse
|
5
|
Roczkowsky A, Chan BYH, Lee TYT, Mahmud Z, Hartley B, Julien O, Armanious G, Young HS, Schulz R. Myocardial MMP-2 contributes to SERCA2a proteolysis during cardiac ischaemia-reperfusion injury. Cardiovasc Res 2020; 116:1021-1031. [PMID: 31373602 DOI: 10.1093/cvr/cvz207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/05/2019] [Accepted: 07/31/2019] [Indexed: 01/06/2023] Open
Abstract
AIMS Matrix metalloproteinase-2 (MMP-2) is a zinc-dependent protease which contributes to cardiac contractile dysfunction when activated during myocardial ischaemia-reperfusion (IR) injury. MMP-2 is localized to several subcellular sites inside cardiac myocytes; however, its role in the sarcoplasmic reticulum (SR) is unknown. The Ca2+ ATPase SERCA2a, which pumps cytosolic Ca2+ into the SR to facilitate muscle relaxation, is degraded in cardiac IR injury; however, the protease responsible for this is unclear. We hypothesized that MMP-2 contributes to cardiac contractile dysfunction by proteolyzing SERCA2a, thereby impairing its activity in IR injury. METHODS AND RESULTS Isolated rat hearts were subjected to IR injury in the presence or absence of the selective MMP inhibitor ARP-100, or perfused aerobically as a control. Inhibition of MMP activity with ARP-100 significantly improved the recovery of cardiac mechanical function and prevented the increase of a 70 kDa SERCA2a degradation fragment following IR injury, although 110 kDa SERCA2a and phospholamban levels appeared unchanged. Electrophoresis of IR heart samples followed by LC-MS/MS confirmed the presence of a SERCA2a fragment of ∼70 kDa. MMP-2 activity co-purified with SR-enriched microsomes prepared from the isolated rat hearts. Endogenous SERCA2a in SR-enriched microsomes was proteolyzed to ∼70 kDa products when incubated in vitro with exogenous MMP-2. MMP-2 also cleaved purified porcine SERCA2a in vitro. SERCA activity in SR-enriched microsomes was decreased by IR injury; however, this was not prevented with ARP-100. CONCLUSION This study shows that MMP-2 activity is found in SR-enriched microsomes from heart muscle and that SERCA2a is proteolyzed by MMP-2. The cardioprotective actions of MMP inhibition in myocardial IR injury may include the prevention of SERCA2a degradation.
Collapse
Affiliation(s)
- Andrej Roczkowsky
- Department of Pediatrics, University of Alberta, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, Edmonton, AB T6G 2S2, Canada.,Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Brandon Y H Chan
- Department of Pediatrics, University of Alberta, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, Edmonton, AB T6G 2S2, Canada.,Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Tim Y T Lee
- Department of Pediatrics, University of Alberta, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, Edmonton, AB T6G 2S2, Canada.,Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Bridgette Hartley
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Gareth Armanious
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Department of Pediatrics, University of Alberta, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, Edmonton, AB T6G 2S2, Canada.,Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
6
|
Manning JR, Wijeratne AB, Oloizia BB, Zhang Y, Greis KD, Schultz JEJ. Phosphoproteomic analysis identifies phospho-Threonine-17 site of phospholamban important in low molecular weight isoform of fibroblast growth factor 2-induced protection against post-ischemic cardiac dysfunction. J Mol Cell Cardiol 2020; 148:1-14. [PMID: 32853649 DOI: 10.1016/j.yjmcc.2020.08.006] [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: 02/05/2020] [Revised: 07/04/2020] [Accepted: 08/09/2020] [Indexed: 10/23/2022]
Abstract
RATIONALE Among its many biological roles, fibroblast growth factor 2 (FGF2) protects the heart from dysfunction and damage associated with an ischemic attack. Our laboratory demonstrated that its protection against myocardial dysfunction occurs by the low molecular weight (LMW) isoform of FGF2, while the high molecular weight (HMW) isoforms are associated with a worsening in post-ischemic recovery of cardiac function. LMW FGF2-mediated cardioprotection is facilitated by activation of multiple kinases, including PKCalpha, PKCepsilon, and ERK, and inhibition of p38 and JNK. OBJECTIVE Yet, the substrates of those kinases associated with LMW FGF2-induced cardioprotection against myocardial dysfunction remain to be elucidated. METHODS AND RESULTS To identify substrates in LMW FGF2 improvement of post-ischemic cardiac function, mouse hearts expressing only LMW FGF2 were subjected to ischemia-reperfusion (I/R) injury and analyzed by a mass spectrometry (MS)-based quantitative phosphoproteomic strategy. MS analysis identified 50 phosphorylation sites from 7 sarcoendoplasmic reticulum (SR) proteins that were significantly altered in I/R-treated hearts only expressing LMW FGF2 compared to those hearts lacking FGF2. One of those phosphorylated SR proteins identified was phospholamban (PLB), which exhibited rapid, increased phosphorylation at Threonine-17 (Thr17) after I/R in hearts expressing only LMW FGF2; this was further validated using Selected Reaction Monitoring-based MS workflow. To demonstrate a mechanistic role of phospho-Thr17 PLB in LMW FGF2-mediated cardioprotection, hearts only expressing LMW FGF2 and those expressing only LMW FGF2 with a mutant PLB lacking phosphorylatable Thr17 (Thr17Ala PLB) were subjected to I/R. Hearts only expressing LMW FGF2 showed significantly improved recovery of cardiac function following I/R (p < 0.05), and this functional improvement was significantly abrogated in hearts expressing LMW FGF2 and Thr17Ala PLB (p < 0.05). CONCLUSION The findings indicate that LMW FGF2 modulates intracellular calcium handling/cycling via regulatory changes in SR proteins essential for recovery from I/R injury, and thereby protects the heart from post-ischemic cardiac dysfunction.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Aruna B Wijeratne
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Brian B Oloizia
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Yu Zhang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America
| | - Jo El J Schultz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States of America.
| |
Collapse
|
7
|
Liu Z, Zhang Y, Qiu C, Zhu H, Pan S, Jia H, Kang H, Guan G, Hui R, Zhu L, Wang J. Diabetes mellitus exacerbates post-myocardial infarction heart failure by reducing sarcolipin promoter methylation. ESC Heart Fail 2020; 7:1935-1948. [PMID: 32525286 PMCID: PMC7373908 DOI: 10.1002/ehf2.12789] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Aims Sarcolipin (SLN) is a key regulator of sarcoplasmic reticulum calcium‐ATPase (SERCA)2a, which handles intracellular calcium re‐uptake. This study was aimed to investigate the involvement of SLN in post‐myocardial infarction (MI) heart failure (HF) in diabetes. Methods and results Diabetes/MI rat models were established. Altered SLN expression in diabetic hearts was screened out by microarray. A myocardiotropic viral vector was used to deliver siRNA to silence SLN. DNA methylation was evaluated by bisulfite sequencing. Cardiac functions were evaluated by invasive haemodynamic examinations. The SERCA2a activity, cytoplasmic calcium concentration ([Ca2+]i), calcium spark, and myocyte contraction were detected. Correlation between HF and diabetes was analysed in a cohort consisted of 101 ST‐segment elevated myocardial infarction (STEMI) patients between 2017 and 2019 [53.54 ± 4.64 years old; 61.4% male gender; HbA1c% 6.15 ± 2.00; and left ventricular ejection fraction (LVEF%) 40.64 ± 3.20%]. SLN expression was evaluated in left ventricular tissue sample from six STEMI patients complicated with diabetes and six STEMI patients without diabetes. Expressions of DNA methyltransferase 1a and DNA methyltransferase 3 were reduced in diabetic hearts, leading to down‐regulation of SLN promoter methylation, resulting in increased SLN expression in rats. Impaired heart systolic functions were found in experimental diabetic MI rats, which were attenuated by SLN silencing. SERCA2a activity reduction and [Ca2+]i elevation were attenuated by SLN silencing in diabetic animal hearts and high‐glucose incubated primary myocytes. SLN silencing suppressed calcium sparks and improved contraction and sarcoplasmic reticulum calcium re‐uptake in high‐glucose incubated primary myocytes. Expression of SLN was up‐regulated in LV sampled from STEMI patients complicated with diabetes compared with non‐diabetic ones (P < 0.05). LVEF% was reduced in STEMI patients complicated with diabetes compared with non‐diabetic ones (P < 0.01). HbA1c% and LVEF% was related (r = −0.218, P = 0.028). Increased HbA1c% was correlated with reduced LVEF% after adjustment for age, sex, body mass index, cigarette smoking, creatinine, UA, low density lipoprotein, K+, Na+, and troponin I (adjusted odds ration = 0.75, 95% confidence interval 0.62–0.90, P = 0.002). Conclusions Diabetes increases the vulnerability of STEMI patients to post‐MI HF by down‐regulating SLN promoter methylation, which further regulates SERCA2a activity via increasing cardiac SLN expression.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Chuan Qiu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Haitao Zhu
- Department of Pediatrics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Shuo Pan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Hao Jia
- International Medical Services, Affiliated Hospital of Northwest University, Xi'an, China
| | - Hongyan Kang
- Department of Cardiology, Heyang County People's Hospital, Weinan, China
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Rutai Hui
- Department of Cardiology, Fuwai Hospital, National Center of Cardiovascular Diseases, Beijing, China
| | - Ling Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| |
Collapse
|
8
|
Menezes-Rodrigues FS, Tavares JGP, Vasques ER, Errante PR, Araújo EAD, Pires-Oliveira M, Scorza CA, Scorza FA, Taha MO, Caricati-Neto A. Cardioprotective effects of pharmacological blockade of the mitochondrial calcium uniporter on myocardial ischemia-reperfusion injury. Acta Cir Bras 2020; 35:e202000306. [PMID: 32692797 PMCID: PMC7251977 DOI: 10.1590/s0102-865020200030000006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/22/2020] [Indexed: 05/13/2023] Open
Abstract
PURPOSE To evaluate whether the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of mitochondrial Ca2+ uniporter (MCU) protects the myocardium against injuries caused by cardiac ischemia and reperfusion (CIR). METHODS CIR was induced in adult male Wistar rats (300-350 g) by occlusion of the left anterior descendent coronary artery (10 min), followed by reperfusion (120 min). Rats were treated with different doses of MCU blocker ruthenium red (RuR), administered 5 min before ischemia or reperfusion. RESULTS In untreated rats, the incidences of ventricular arrhythmias (VA), atrioventricular block (AVB) and the lethality (LET) induced by CIR were 85%, 79% and 70%, respectively. In rats treated with RuR before ischemia, the incidences of VA, AVB and LET were significantly reduced to 62%, 25% and 25%, respectively. In rats treated with RuR after ischemia, the incidences of VA, AVB and LET were significantly reduced to 50%, 25% and 25%, respectively. CONCLUSION The significant reduction of the incidence of CIR-induced VA, AVB and LET produced by the treatment with RuR indicates that the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of MCU can protect the myocardium against injuries caused by CIR.
Collapse
|
9
|
Horowitz JD, Chong CR. Matrix metalloproteinase-2 activation: critical to myocardial contractile dysfunction following ischaemia-reperfusion. Cardiovasc Res 2020; 116:876-878. [PMID: 31800010 DOI: 10.1093/cvr/cvz271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- John D Horowitz
- Basil Hetzel Institute for Translational Research, The Queen Elizabeth Hospital, Woodville Road, Woodville, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Cher-Rin Chong
- Basil Hetzel Institute for Translational Research, The Queen Elizabeth Hospital, Woodville Road, Woodville, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
10
|
Torres MJ, McLaughlin KL, Renegar RH, Valsaraj S, Whitehurst KS, Sharaf OM, Sharma UM, Horton JL, Sarathy B, Parks JC, Brault JJ, Fisher-Wellman KH, Neufer PD, Virag JAI. Intracardiac administration of ephrinA1-Fc preserves mitochondrial bioenergetics during acute ischemia/reperfusion injury. Life Sci 2019; 239:117053. [PMID: 31733316 DOI: 10.1016/j.lfs.2019.117053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022]
Abstract
AIMS Intracardiac injection of recombinant EphrinA1-Fc immediately following coronary artery ligation in mice reduces infarct size in both reperfused and non-reperfused myocardium, but the cellular alterations behind this phenomenon remain unknown. MAIN METHODS Herein, 10 wk-old B6129SF2/J male mice were exposed to acute ischemia/reperfusion (30minI/24hrsR) injury immediately followed by intracardiac injection of either EphrinA1-Fc or IgG-Fc. After 24 h of reperfusion, sections of the infarct margin in the left ventricle were imaged via transmission electron microscopy, and mitochondrial function was assessed in both permeabilized fibers and isolated mitochondria, to examine mitochondrial structure, function, and energetics in the early stages of repair. KEY FINDINGS At a structural level, EphrinA1-Fc administration prevented the I/R-induced loss of sarcomere alignment and mitochondrial organization along the Z disks, as well as disorganization of the cristae and loss of inter-mitochondrial junctions. With respect to bioenergetics, loss of respiratory function induced by I/R was prevented by EphrinA1-Fc. Preservation of cardiac bioenergetics was not due to changes in mitochondrial JH2O2 emitting potential, membrane potential, ADP affinity, efficiency of ATP production, or activity of the main dehydrogenase enzymes, suggesting that EphrinA1-Fc indirectly maintains respiratory function via preservation of the mitochondrial network. Moreover, these protective effects were lost in isolated mitochondria, further emphasizing the importance of the intact cardiomyocyte ultrastructure in mitochondrial energetics. SIGNIFICANCE Collectively, these data suggest that intracardiac injection of EphrinA1-Fc protects cardiac function by preserving cardiomyocyte structure and mitochondrial bioenergetics, thus emerging as a potential therapeutic strategy in I/R injury.
Collapse
Affiliation(s)
- Maria J Torres
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA
| | - Kelsey L McLaughlin
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Randall H Renegar
- Dept of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Smrithi Valsaraj
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - K'Shylah S Whitehurst
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Omar M Sharaf
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Uma M Sharma
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Julie L Horton
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Brinda Sarathy
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Justin C Parks
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Jeffrey J Brault
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Dept of Kinesiology, College of Health and Human Performance, East Carolina University, Greenville, NC, 27834, USA
| | - Kelsey H Fisher-Wellman
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA; Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Jitka A I Virag
- Dept of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
11
|
Lock MC, Darby JRT, Soo JY, Brooks DA, Perumal SR, Selvanayagam JB, Seed M, Macgowan CK, Porrello ER, Tellam RL, Morrison JL. Differential Response to Injury in Fetal and Adolescent Sheep Hearts in the Immediate Post-myocardial Infarction Period. Front Physiol 2019; 10:208. [PMID: 30890961 PMCID: PMC6412108 DOI: 10.3389/fphys.2019.00208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Aim: Characterizing the response to myocardial infarction (MI) in the regenerative sheep fetus heart compared to the post-natal non-regenerative adolescent heart may reveal key morphological and molecular differences that equate to the response to MI in humans. We hypothesized that the immediate response to injury in (a) infarct compared with sham, and (b) infarct, border, and remote tissue, in the fetal sheep heart would be fundamentally different to the adolescent, allowing for repair after damage. Methods: We used a sheep model of MI induced by ligating the left anterior descending coronary artery. Surgery was performed on fetuses (105 days) and adolescent sheep (6 months). Sheep were randomly separated into MI (n = 5) or Sham (n = 5) surgery groups at both ages. We used magnetic resonance imaging (MRI), histological/immunohistochemical staining, and qRT-PCR to assess the morphological and molecular differences between the different age groups in response to infarction. Results: Magnetic resonance imaging showed no difference in fetuses for key functional parameters; however there was a significant decrease in left ventricular ejection fraction and cardiac output in the adolescent sheep heart at 3 days post-infarction. There was no significant difference in functional parameters between MRI sessions at Day 0 and Day 3 after surgery. Expression of genes involved in glucose transport and fatty acid metabolism, inflammatory cytokines as well as growth factors and cell cycle regulators remained largely unchanged in the infarcted compared to sham ventricular tissue in the fetus, but were significantly dysregulated in the adolescent sheep. Different cardiac tissue region-specific gene expression profiles were observed between the fetal and adolescent sheep. Conclusion: Fetuses demonstrated a resistance to cardiac damage not observed in the adolescent animals. The manipulation of specific gene expression profiles to a fetal-like state may provide a therapeutic strategy to treat patients following an infarction.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sunthara Rajan Perumal
- Preclinical, Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Joseph B Selvanayagam
- Cardiac Imaging Research Group, Department of Heart Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Mike Seed
- The Hospital for Sick Children, Division of Cardiology, Toronto, ON, Canada
| | | | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
12
|
Yu S, Guo Y, Zhang W, Zheng L, Ren J, Jin J, Yu B, Zhang Y, Wang H, Zhang Y. [Effects of propofol pretreatment on myocardial cell apoptosis and SERCA2 expression in rats with hepatic ischemia/reperfusion]. Rev Bras Anestesiol 2018; 68:591-596. [PMID: 30195630 DOI: 10.1016/j.bjan.2018.06.003] [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: 09/26/2016] [Revised: 06/03/2018] [Accepted: 06/15/2018] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Hepatic ischemia-reperfusion injury is a common pathophysiological process in liver surgery. Whether Propofol can reduce myocardial ischemia-reperfusion injury induced by hepatic ischemia-reperfusion injury in rats, together with related mechanisms, still needs further studies. OBJECTIVE To investigate if propofol would protect the myocardial cells from apoptosis with hepatic ischemia-reperfusion injury. METHODS Male Sprague-Dawley rats (n=18) were randomly allocated into three groups: Sham Group (Group S, n=6), Hepatic Ischemia-reperfusion Injury Group (Group IR, n=6) and Propofol Group (Group P, n=6). Group S was only subjected to laparotomy. Group IR was attained by ischemia for 30min and reperfusion for 4h. Group P was subjected identical insult as in Group IR with the administration of propofol started 10min before ischemia with 120mg.kg-1, following by continuous infusion at 20mg.kg-1.h-1. Cell apoptosis was examined by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. Endoplasmic reticulum Ca2+-ATPase2 (SERCA2) and cysteine-containing aspartic acid cleaved-caspase3 (cleaved-caspase3) were assayed by western blot and Altimeter polymerase chain reaction. RESULTS Apoptosis rate was increased, with mRNA and protein of SERCA2 down-regulated and cleaved-caspase3 up-regulated in Group IR compared with Group S (p<0.01). Apoptosis rate was decreased, with mRNA and protein of SERCA2 up-regulated and cleaved-caspase3 down-regulated in Group P compared with Group IR (p<0.01). CONCLUSIONS Propofol can reduce hepatic ischemia-reperfusion injury-induced myocardial cell apoptosis, meanwhile, can up-regulate mRNA and protein of SERCA2 in rats.
Collapse
Affiliation(s)
- Shuzhen Yu
- Shanxi Provincial People's Hospital, Department of Anesthesiology, Taiyuan, China
| | - Yongqing Guo
- Shanxi Provincial People's Hospital, Department of Anesthesiology, Taiyuan, China.
| | - Weiwei Zhang
- Shanxi Provincial People's Hospital, Department of Anesthesiology, Taiyuan, China
| | - Lina Zheng
- Shanxi Provincial People's Hospital, Department of Anesthesiology, Taiyuan, China
| | - Junming Ren
- Shanxi Medical University, Department of Biochemistry, Taiyuan, China
| | - Jianmin Jin
- Shanxi Provincial People's Hospital, Department of Anesthesiology, Taiyuan, China
| | - Baofeng Yu
- Shanxi Medical University, Department of Biochemistry, Taiyuan, China
| | - Yu Zhang
- Shanxi People's Hospital, Department of General Surgery, Taiyuan, China
| | - Hao Wang
- Shanxi People's Hospital, Department of General Surgery, Taiyuan, China
| | - Yuhong Zhang
- Shanxi People's Hospital, Department of General Surgery, Taiyuan, China
| |
Collapse
|
13
|
O'Shea KM, Ananthakrishnan R, Li Q, Quadri N, Thiagarajan D, Sreejit G, Wang L, Zirpoli H, Aranda JF, Alberts AS, Schmidt AM, Ramasamy R. The Formin, DIAPH1, is a Key Modulator of Myocardial Ischemia/Reperfusion Injury. EBioMedicine 2017; 26:165-174. [PMID: 29239839 PMCID: PMC5832565 DOI: 10.1016/j.ebiom.2017.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/16/2017] [Accepted: 11/16/2017] [Indexed: 01/13/2023] Open
Abstract
The biochemical, ionic, and signaling changes that occur within cardiomyocytes subjected to ischemia are exacerbated by reperfusion; however, the precise mechanisms mediating myocardial ischemia/reperfusion (I/R) injury have not been fully elucidated. The receptor for advanced glycation end-products (RAGE) regulates the cellular response to cardiac tissue damage in I/R, an effect potentially mediated by the binding of the RAGE cytoplasmic domain to the diaphanous-related formin, DIAPH1. The aim of this study was to investigate the role of DIAPH1 in the physiological response to experimental myocardial I/R in mice. After subjecting wild-type mice to experimental I/R, myocardial DIAPH1 expression was increased, an effect that was echoed following hypoxia/reoxygenation (H/R) in H9C2 and AC16 cells. Further, compared to wild-type mice, genetic deletion of Diaph1 reduced infarct size and improved contractile function after I/R. Silencing Diaph1 in H9C2 cells subjected to H/R downregulated actin polymerization and serum response factor-regulated gene expression. Importantly, these changes led to increased expression of sarcoplasmic reticulum Ca2+ ATPase and reduced expression of the sodium calcium exchanger. This work demonstrates that DIAPH1 is required for the myocardial response to I/R, and that targeting DIAPH1 may represent an adjunctive approach for myocardial salvage after acute infarction.
Collapse
Affiliation(s)
- Karen M O'Shea
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Radha Ananthakrishnan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Qing Li
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Devi Thiagarajan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Gopalkrishna Sreejit
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Lingjie Wang
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Hylde Zirpoli
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Juan Francisco Aranda
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Arthur S Alberts
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
14
|
Wang S, Ding L, Ji H, Xu Z, Liu Q, Zheng Y. The Role of p38 MAPK in the Development of Diabetic Cardiomyopathy. Int J Mol Sci 2016; 17:ijms17071037. [PMID: 27376265 PMCID: PMC4964413 DOI: 10.3390/ijms17071037] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a major complication of diabetes that contributes to an increase in mortality. A number of mechanisms potentially explain the development of DCM including oxidative stress, inflammation and extracellular fibrosis. Mitogen-activated protein kinase (MAPK)-mediated signaling pathways are common among these pathogenic responses. Among the diverse array of kinases, extensive attention has been given to p38 MAPK due to its capacity for promoting or inhibiting the translation of target genes. Growing evidence has indicated that p38 MAPK is aberrantly expressed in the cardiovascular system, including the heart, under both experimental and clinical diabetic conditions and, furthermore, inhibition of p38 MAPK activation in transgenic animal model or with its pharmacologic inhibitor significantly prevents the development of DCM, implicating p38 MAPK as a novel diagnostic indicator and therapeutic target for DCM. This review summarizes our current knowledge base to provide an overview of the impact of p38 MAPK signaling in diabetes-induced cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Shudong Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Lijuan Ding
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Honglei Ji
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zheng Xu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Quan Liu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yang Zheng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
15
|
Abstract
Eukaryotic cells have been confronted throughout their evolution with potentially lethal plasma membrane injuries, including those caused by osmotic stress, by infection from bacterial toxins and parasites, and by mechanical and ischemic stress. The wounded cell can survive if a rapid repair response is mounted that restores boundary integrity. Calcium has been identified as the key trigger to activate an effective membrane repair response that utilizes exocytosis and endocytosis to repair a membrane tear, or remove a membrane pore. We here review what is known about the cellular and molecular mechanisms of membrane repair, with particular emphasis on the relevance of repair as it relates to disease pathologies. Collective evidence reveals membrane repair employs primitive yet robust molecular machinery, such as vesicle fusion and contractile rings, processes evolutionarily honed for simplicity and success. Yet to be fully understood is whether core membrane repair machinery exists in all cells, or whether evolutionary adaptation has resulted in multiple compensatory repair pathways that specialize in different tissues and cells within our body.
Collapse
Affiliation(s)
- Sandra T Cooper
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| | - Paul L McNeil
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
16
|
Talukder MAH, Elnakish MT, Yang F, Nishijima Y, Alhaj MA, Velayutham M, Hassanain HH, Zweier JL. Cardiomyocyte-specific overexpression of an active form of Rac predisposes the heart to increased myocardial stunning and ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2013; 304:H294-H302. [PMID: 23161879 PMCID: PMC3543663 DOI: 10.1152/ajpheart.00367.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022]
Abstract
The GTP-binding protein Rac regulates diverse cellular functions including activation of NADPH oxidase, a major source of superoxide production (O(2)(·-)). Rac1-mediated NADPH oxidase activation is increased after myocardial infarction (MI) and heart failure both in animals and humans; however, the impact of increased myocardial Rac on impending ischemia-reperfusion (I/R) is unknown. A novel transgenic mouse model with cardiac-specific overexpression of constitutively active mutant form of Zea maize Rac D (ZmRacD) gene has been reported with increased myocardial Rac-GTPase activity and O(2)(·-) generation. The goal of the present study was to determine signaling pathways related to increased myocardial ZmRacD and to what extent hearts with increased ZmRacD proteins are susceptible to I/R injury. The effect of myocardial I/R was examined in young adult wild-type (WT) and ZmRacD transgenic (TG) mice. In vitro reversible myocardial I/R for postischemic cardiac function and in vivo regional myocardial I/R for MI were performed. Following 20-min global ischemia and 45-min reperfusion, postischemic cardiac contractile function and heart rate were significantly reduced in TG hearts compared with WT hearts. Importantly, acute regional myocardial I/R (30-min ischemia and 24-h reperfusion) caused significantly larger MI in TG mice compared with WT mice. Western blot analysis of cardiac homogenates revealed that increased myocardial ZmRacD gene expression is associated with concomitant increased levels of NADPH oxidase subunit gp91(phox), O(2)(·-), and P(21)-activated kinase. Thus these findings provide direct evidence that increased levels of active myocardial Rac renders the heart susceptible to increased postischemic contractile dysfunction and MI following acute I/R.
Collapse
Affiliation(s)
- M A Hassan Talukder
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Nielsen TT, Støttrup NB, Løfgren B, Bøtker HE. Metabolic fingerprint of ischaemic cardioprotection: importance of the malate-aspartate shuttle. Cardiovasc Res 2011; 91:382-91. [PMID: 21349875 DOI: 10.1093/cvr/cvr051] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The convergence of cardioprotective intracellular signalling pathways to modulate mitochondrial function as an end-target of cytoprotective stimuli is well described. However, our understanding of whether the complementary changes in mitochondrial energy metabolism are secondary responses or inherent mechanisms of ischaemic cardioprotection remains incomplete. In the heart, the malate-aspartate shuttle (MAS) constitutes the primary metabolic pathway for transfer of reducing equivalents from the cytosol into the mitochondria for oxidation. The flux of MAS is tightly linked to the flux of the tricarboxylic acid cycle and the electron transport chain, partly by the amino acid l-glutamate. In addition, emerging evidence suggests the MAS is an important regulator of cytosolic and mitochondrial calcium homeostasis. In the isolated rat heart, inhibition of MAS during ischaemia and early reperfusion by the aminotransferase inhibitor aminooxyacetate induces infarct limitation, improves haemodynamic responses, and modulates glucose metabolism, analogous to effects observed in classical ischaemic preconditioning. On the basis of these findings, the mechanisms through which MAS preserves mitochondrial function and cell survival are reviewed. We conclude that the available evidence is supportive of a down-regulation of mitochondrial respiration during lethal ischaemia with a gradual 'wake-up' during reperfusion as a pivotal feature of ischaemic cardioprotection. Finally, comments on modulating myocardial energy metabolism by the cardioprotective amino acids glutamate and glutamine are given.
Collapse
Affiliation(s)
- Torsten Toftegaard Nielsen
- Department of Cardiology, Skejby Hospital, Aarhus University Hospital, Brendstrupgaardsvej 100, Aarhus N, Denmark.
| | | | | | | |
Collapse
|
18
|
Talukder MAH, Yang F, Nishijima Y, Chen CA, Xie L, Mahamud SD, Kalyanasundaram A, Bonagura JD, Periasamy M, Zweier JL. Detrimental effects of thyroid hormone analog DITPA in the mouse heart: increased mortality with in vivo acute myocardial ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2011; 300:H702-H711. [PMID: 21131480 PMCID: PMC3044051 DOI: 10.1152/ajpheart.00514.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 11/23/2010] [Indexed: 11/22/2022]
Abstract
There is emerging evidence that treatment with thyroid hormone (TH) can improve postischemic cardiac function. 3,5-Diiodothyropropionic acid (DITPA), a TH analog, has been proposed to be a safer therapeutic agent than TH because of its negligible effects on cardiac metabolism and heart rate. However, conflicting results have been reported for the cardiac effects of DITPA. Importantly, recent clinical trials demonstrated no symptomatic benefit in patients with DITPA despite some improved hemodynamic and metabolic parameters. To address these issues, dose-dependent effects of DITPA were investigated in mice for baseline cardiovascular effects and postischemic myocardial function and/or salvage. Mice were treated with subcutaneous DITPA at 0.937, 1.875, 3.75, or 7.5 mg·kg(-1)·day(-1) for 7 days, and the results were compared with untreated mice for ex vivo and/or in vivo myocardial ischemia-reperfusion (I/R). DITPA had no effects on baseline body temperature, body weight, or heart rate; however, it mildly increased blood pressure. In isolated hearts, baseline contractile function was significantly impaired in DITPA-pretreated mice; however, postischemic recovery was comparable between untreated and DITPA-treated groups. In vivo baseline cardiac parameters were significantly affected by DITPA, with increased ventricular dimensions and decreased contractile function. Importantly, DITPA-treated mice demonstrated high prevalence of fatal cardiac rhythm abnormalities during in vivo ischemia and/or reperfusion. There were no improvements in myocardial infarction and postischemic fractional shortening with DITPA. Myocardial sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA), phospholamban (PLB), and heat shock protein (HSP) levels remained unchanged with DITPA treatment. Thus DITPA administration impairs baseline cardiac parameters in mice and can be fatal during in vivo acute myocardial I/R.
Collapse
Affiliation(s)
- M A Hassan Talukder
- Davis Heart and Lung Research Institute, Department of Internal Medicine, Ohio State University College of Medicine, 473 West 12th Ave., Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Talukder MH, Zweier JL, Periasamy M. Targeting calcium transport in ischaemic heart disease. Cardiovasc Res 2009; 84:345-52. [PMID: 19640931 PMCID: PMC2777954 DOI: 10.1093/cvr/cvp264] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 07/14/2009] [Accepted: 07/20/2009] [Indexed: 01/14/2023] Open
Abstract
Ischaemic heart disease (IHD) is the leading cause of morbidity and mortality worldwide. While timely reperfusion of acutely ischaemic myocardium is essential for myocardial salvage, it leads to a unique type of injury known as 'myocardial ischaemia/reperfusion (I/R) injury'. Growing evidence suggests that a defect in myocardial Ca(2+) transport system with cytosolic Ca(2+) overload is a major contributor to myocardial I/R injury. Progress in molecular genetics and medicine in past years has clearly demonstrated that modulation of Ca(2+) handling pathways in IHD could be cardioprotective. The potential benefits of these strategies in limiting I/R injury are vast, and the time is right for challenging in vivo systemic work both at pre-clinical and clinical levels.
Collapse
Affiliation(s)
- M.A. Hassan Talukder
- Davis Heart and Lung Institute and The Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Jay L. Zweier
- Davis Heart and Lung Institute and The Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, 304 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA
| | - Muthu Periasamy
- Davis Heart and Lung Institute and The Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, 304 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA
| |
Collapse
|