1
|
Aubry A, Kebbe M, Naud P, Villeneuve L, Leblanc CA, Calderone A. Nestin (+)- and Nestin (-)-Ventricular Cardiomyocytes Reenter the Cell Cycle In Vitro but Are Reciprocally Regulated in the Partial Apex-Resected 7-Day Neonatal Rat Heart. J Cell Physiol 2025; 240:e70040. [PMID: 40275768 PMCID: PMC12022726 DOI: 10.1002/jcp.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
The 1-day-old neonatal rat heart contains two subpopulations of ventricular cardiomyocytes (NNVMs) that reenter the cell cycle in vitro and in vivo distinguished by the absence or de novo expression of the intermediate filament protein nestin. Furthermore, de novo nestin expression in NNVMs directly facilitated cell cycle reentry and elicited a morphological migratory phenotype. Previous studies have reported that ventricular cardiomyocytes failed to reenter the cell cycle following damage to the 7-day-old rodent heart. The present study tested the hypothesis that cell cycle reentry of one or both of the NNVM subpopulations of 7-day-old neonatal rat pups was compromised in vitro and/or in vivo following cardiac damage. Three-day treatment of 7-day-old NNVMs with the protein kinase C activator phorbol 12,13-dibutyrate and the serine/threonine p38α/β MAPK kinase inhibitor SB203580 facilitated cell cycle reentry into the S phase and G2-M phase of the cell cycle. Two distinct subpopulations of 7-day NNVMs reentered the cell cycle, and the predominant subpopulation was distinguished by de novo nestin expression. Three days following the sham-operation of 7-day-old neonatal rat hearts, cell cycle reentry was detected exclusively in NNVMs lacking nestin expression. Partial apex resection of 7-day-old neonatal rat hearts led to the de novo appearance of nestin(+)-NNVMs preferentially bordering the damaged region and a subpopulation reentered the S-phase and G2-M phase of the cell cycle in the absence of p38α/β MAPK inhibition. By contrast, cell cycle reentry of nestin(-)-NNVMs identified adjacent to the apex-resected region was significantly reduced. These data highlight the disparate in vivo regulation of the two subpopulations of NNVMs following damaged to the 7-day-old neonatal rat heart and reaffirm the premise that targeting the subpopulation of nestin(+)-ventricular cardiomyocytes identified in the ischemically damaged adult mammalian heart represents a plausible first step to initiate cell cycle reentry.
Collapse
Affiliation(s)
- Adrien Aubry
- Montreal Heart InstituteUniversité de MontréalMontréalQuébecCanada
- Département de Pharmacologie et PhysiologieUniversité de MontréalMontréalQuébecCanada
| | - Mariana Kebbe
- Montreal Heart InstituteUniversité de MontréalMontréalQuébecCanada
- Département de Pharmacologie et PhysiologieUniversité de MontréalMontréalQuébecCanada
| | - Patrice Naud
- Montreal Heart InstituteUniversité de MontréalMontréalQuébecCanada
| | - Louis Villeneuve
- Montreal Heart InstituteUniversité de MontréalMontréalQuébecCanada
| | | | - Angelino Calderone
- Montreal Heart InstituteUniversité de MontréalMontréalQuébecCanada
- Département de Pharmacologie et PhysiologieUniversité de MontréalMontréalQuébecCanada
| |
Collapse
|
2
|
Priya A, Mol N, Singh AK, Aditya AK, Ray AK. "Unveiling the impacts of climatic cold events on the cardiovascular health in animal models". THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 971:179028. [PMID: 40073773 DOI: 10.1016/j.scitotenv.2025.179028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Climate change is increasingly driving extreme weather events, leading to drastic temperature fluctuations worldwide. While overall temperatures rise, many regions are simultaneously experiencing severe cold spells that threaten the health of human populations, especially to vulnerable populations including the elderly and those with pre-existing conditions. Exposure to cold stress triggers significant physiological and biochemical disruptions. As cardiovascular diseases (CVDs) rank among the leading causes of global morbidity and mortality, the exacerbation of these conditions by cold exposure underscores critical public health challenges. The complex pathophysiological processes in cold-induced CVDs require careful analysis at an organ-system level, making animal models an ideal tool for replicating human physiological and molecular responses in a controlled environment. However, a detailed mechanism linking cold exposure and cardiovascular dysfunction remains incompletely understood, particularly in the context of animal models. Therefore, this comprehensive review aims to address and analyze from traditional rodent models to less conventional ruminants, broilers, canines, and primate animal models to understand cold stress-induced CVDs, with an extensive account of the potential molecular mechanisms and pathways such as oxidative stress, inflammation, vasomotor dysfunction, and apoptosis, along with emerging roles of cold shock proteins (CSPs), etc. We also delve into various potential therapeutic approaches and preventive measures in cold stress conditions. In conclusion, this review is the first to comprehensively address the underexplored cardiovascular complications arising from cold stress and their underlying mechanisms, particularly using animal models. Furthermore, it provides a foundation for advancing the development of more effective and targeted therapies through translational research.
Collapse
Affiliation(s)
- Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Nidhi Mol
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Centre, Mathura, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
3
|
Liu H, Magaye R, Kaye DM, Wang BH. Heart failure with preserved ejection fraction: The role of inflammation. Eur J Pharmacol 2024; 980:176858. [PMID: 39074526 DOI: 10.1016/j.ejphar.2024.176858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Heart failure (HF) is a debilitating clinical syndrome affecting 64.3 million patients worldwide. More than 50% of HF cases are attributed to HF with preserved ejection fraction (HFpEF), an entity growing in prevalence and mortality. Although recent breakthroughs reveal the prognostic benefits of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in HFpEF, there is still a lack of effective pharmacological therapy available. This highlights a major gap in medical knowledge that must be addressed. Current evidence attributes HFpEF pathogenesis to an interplay between cardiometabolic comorbidities, inflammation, and renin-angiotensin-aldosterone-system (RAAS) activation, leading to cardiac remodelling and diastolic dysfunction. However, conventional RAAS blockade has demonstrated limited benefits in HFpEF, which emphasises that alternative therapeutic targets should be explored. Presently, there is limited literature examining the use of anti-inflammatory HFpEF therapies despite growing evidence supporting its importance in disease progression. Hence, this review aims to explore current perspectives on HFpEF pathogenesis, including the importance of inflammation-driven cardiac remodelling and the therapeutic potential of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hongyi Liu
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Ruth Magaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - David M Kaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Bing H Wang
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
4
|
Ikushima A, Ishimura T, Mori KP, Yamada H, Sugioka S, Ishii A, Toda N, Ohno S, Kato Y, Handa T, Yanagita M, Yokoi H. Deletion of p38 MAPK in macrophages ameliorates peritoneal fibrosis and inflammation in peritoneal dialysis. Sci Rep 2024; 14:21220. [PMID: 39261560 PMCID: PMC11391064 DOI: 10.1038/s41598-024-71859-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
One of the most common causes of peritoneal dialysis withdrawal is ultrafiltration failure which is characterized by peritoneal membrane thickening and fibrosis. Although previous studies have demonstrated the inhibitory effect of p38 MAPK inhibitors on peritoneal fibrosis in mice, it was unclear which specific cells contribute to peritoneal fibrosis. To investigate the role of p38 MAPK in peritoneal fibrosis more precisely, we examined the expression of p38 MAPK in human peritoneum and generated systemic inducible p38 MAPK knockout mice and macrophage-specific p38 MAPK knockout mice. Furthermore, the response to lipopolysaccharide (LPS) was assessed in p38 MAPK-knocked down RAW 264.7 cells to further explore the role of p38 MAPK in macrophages. We found that phosphorylated p38 MAPK levels were increased in the thickened peritoneum of both human and mice. Both chlorhexidine gluconate (CG)-treated systemic inducible and macrophage-specific p38 MAPK knockout mice ameliorated peritoneal thickening, mRNA expression related to inflammation and fibrosis, and the number of αSMA- and MAC-2-positive cells in the peritoneum compared to CG control mice. Reduction of p38 MAPK in RAW 264.7 cells suppressed inflammatory mRNA expression induced by LPS. These findings suggest that p38 MAPK in macrophages plays a critical role in peritoneal inflammation and thickening.
Collapse
Affiliation(s)
- Akie Ikushima
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Takuya Ishimura
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Keita P Mori
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology and Dialysis, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Primary Care & Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sayaka Sugioka
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Akira Ishii
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology, Kansai Electric Power Hospital, Osaka, Japan
| | - Naohiro Toda
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology, Kansai Electric Power Hospital, Osaka, Japan
| | - Shoko Ohno
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Yukiko Kato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Takaya Handa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology and Dialysis, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan.
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan.
| |
Collapse
|
5
|
Li J, Li X, Wang Y, Meng L, Cui W. Zinc: a potential star for regulating peritoneal fibrosis. Front Pharmacol 2024; 15:1436864. [PMID: 39301569 PMCID: PMC11411568 DOI: 10.3389/fphar.2024.1436864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Peritoneal dialysis (PD) is a commonly used renal replacement therapy for patients with end-stage renal disease (ESRD). During PD, the peritoneum (PM), a semi-permeable membrane, is exposed to nonbiocompatible PD solutions. Peritonitis can occur, leading to structural and functional PM disorders, resulting in peritoneal fibrosis and ultrafiltration failure, which are important reasons for patients with ESRD to discontinue PD. Increasing evidence suggests that oxidative stress (OS) plays a key role in the pathogenesis of peritoneal fibrosis. Furthermore, zinc deficiency is often present to a certain extent in patients undergoing PD. As an essential trace element, zinc is also an antioxidant, potentially playing an anti-OS role and slowing down peritoneal fibrosis progression. This study summarises and analyses recent research conducted by domestic and foreign scholars on the possible mechanisms through which zinc prevents peritoneal fibrosis.
Collapse
Affiliation(s)
- Jian Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xinyang Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yangwei Wang
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Lingfei Meng
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Wu Y, Zou Y, Song C, Cao K, Cai K, Chen S, Zhang Z, Geng D, Zhang N, Feng H, Tang M, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of serine/threonine protein kinases in cardiovascular disease and potential therapeutic methods. Biomed Pharmacother 2024; 177:117093. [PMID: 38971012 DOI: 10.1016/j.biopha.2024.117093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
Protein phosphorylation is an important link in a variety of signaling pathways, and most of the important life processes in cells involve protein phosphorylation. Based on the amino acid residues of phosphorylated proteins, protein kinases can be categorized into the following families: serine/threonine protein kinases, tyrosine-specific protein kinases, histidine-specific protein kinases, tryptophan kinases, and aspartate/glutamyl protein kinases. Of all the protein kinases, most are serine/threonine kinases, where serine/threonine protein kinases are protein kinases that catalyze the phosphorylation of serine or threonine residues on target proteins using ATP as a phosphate donor. The current socially accepted classification of serine/threonine kinases is to divide them into seven major groups: protein kinase A, G, C (AGC), CMGC, Calmodulin-dependent protein kinase (CAMK), Casein kinase (CK1), STE, Tyrosine kinase (TKL) and others. After decades of research, a preliminary understanding of the specific classification and respective functions of serine/threonine kinases has entered a new period of exploration. In this paper, we review the literature of the previous years and introduce the specific signaling pathways and related therapeutic modalities played by each of the small protein kinases in the serine/threonine protein kinase family, respectively, in some common cardiovascular system diseases such as heart failure, myocardial infarction, ischemia-reperfusion injury, and diabetic cardiomyopathy. To a certain extent, the current research results, including molecular mechanisms and therapeutic methods, are fully summarized and a systematic report is made for the prevention and treatment of cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China.
| | - Hao Feng
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| |
Collapse
|
7
|
Xu D, Ye B, Lin L, Jin Y, Jiang Y, Zheng Z, Chen Y, Han X, Wang W, Wu G, Zhuang Z, Shan P, Liang G. Carnosol attenuates angiotensin II-induced cardiac remodeling and inflammation via directly binding to p38 and inhibiting p38 activation. Int Immunopharmacol 2024; 134:112143. [PMID: 38692016 DOI: 10.1016/j.intimp.2024.112143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/03/2024]
Abstract
Chronic inflammation is a significant contributor to hypertensive heart failure. Carnosol (Car), primarily derived from the sage plant (Salvia carnosa), exhibits anti-inflammatory properties in a range of systems. Nevertheless, the influence of angiotensin II (Ang II) on cardiac remodeling remains uncharted. Car was shown to protect mice's hearts against Ang II-induced heart damage at dosages of 20 and 40 mg/kg/d. This protection was evident in a concentration-related decrease in the remodeling of the heart and dysfunction. Examination of the transcriptome revealed that the pivotal roles in mediating the protective effects of Car involved inhibiting Ang II-induced inflammation and the activation of the mitogen-activated protein kinase (MAPK) pathway. Furthermore, Car was found to inhibit p38 phosphorylation, therefore reducing the level of inflammation in cultured cardiomyocytes and mouse hearts. This effect was attributed to the direct binding to p38 and inhibition of p38 protein phosphorylation by Car both in vitro and in vivo. In addition, the effects of Car on inflammation were neutralized when p38 was blocked in cardiomyocytes.
Collapse
Affiliation(s)
- Diyun Xu
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou 325800, Zhejiang, China; Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liming Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanhong Jin
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuchen Jiang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhaozheng Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanghao Chen
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Wang
- Department of Neurosurgery, Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China
| | - Gaojun Wu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zaishou Zhuang
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou 325800, Zhejiang, China.
| | - Peiren Shan
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Guang Liang
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou 325800, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Mohammadi K, Shafie D, Ghomashi N, Abdolizadeh A, Sadeghpour M. Kinin-kallikrein system: New perspectives in heart failure. Heart Fail Rev 2024; 29:729-737. [PMID: 38381277 DOI: 10.1007/s10741-024-10393-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Heart failure (HF) is a pervasive clinical challenge characterized by compromised cardiac function and reduced quality of life. The kinin-kallikrein system (KSS), a multifaceted peptide cascade, has garnered substantial attention due to its potential role in HF. Through activation of B1 and/or B2 receptors and downstream signaling, kinins modulate various physiological processes, including inflammation, coagulation, pain, blood pressure control, and vascular permeability. Notably, aberrations in KKS components have been linked to HF risk. The elevation of vasodilatory bradykinin (BK) due to kallikrein activity reduces preload and afterload, while concurrently fostering sodium reabsorption inhibition. However, kallikrein's conversion of prorenin to renin leads to angiotensinsII upregulation, resulting in vasoconstriction and fluid retention, alongside increased immune cell activity that fuels inflammation and cardiac remodeling. Importantly, prolonged KKS activation resulting from volume overload and tissue stretch contributes to cardiac collagen loss. The conventional renin-angiotensin-aldosterone system (RAAS) inhibitors used in HF management may inadvertently intensify KKS activity, exacerbating collagen depletion and cardiac remodeling. It is crucial to balance the KKS's role in acute cardiac damage, which may temporarily enhance function and metabolic parameters against its detrimental long-term effects. Thus, KKS blockade emerges as a promising strategy to impede HF progression. By attenuating the link between immune system function and tissue damage, KKS inhibition can potentially reduce cardiac remodeling and alleviate HF symptoms. However, the nuanced roles of BK in various acute conditions necessitate further investigation into the sustained benefits of kallikrein inhibitors in patients with chronic HF.
Collapse
Affiliation(s)
- Keivan Mohammadi
- Shahid Chamran Heart Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Newsha Ghomashi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Abdolizadeh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Majid Sadeghpour
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
9
|
Ghosh M, Lee J, Burke AN, Strong TA, Sagen J, Pearse DD. Sex Dependent Disparities in the Central Innate Immune Response after Moderate Spinal Cord Contusion in Rat. Cells 2024; 13:645. [PMID: 38607084 PMCID: PMC11011714 DOI: 10.3390/cells13070645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Subacute spinal cord injury (SCI) displays a complex pathophysiology associated with pro-inflammation and ensuing tissue damage. Microglia, the resident innate immune cells of the CNS, in concert with infiltrating macrophages, are the primary contributors to SCI-induced inflammation. However, subpopulations of activated microglia can also possess immunomodulatory activities that are essential for tissue remodeling and repair, including the production of anti-inflammatory cytokines and growth factors that are vital for SCI recovery. Recently, reports have provided convincing evidence that sex-dependent differences exist in how microglia function during CNS pathologies and the extent to which these cells contribute to neurorepair and endogenous recovery. Herein we employed flow cytometry and immunohistochemical methods to characterize the phenotype and population dynamics of activated innate immune cells within the injured spinal cord of age-matched male and female rats within the first week (7 days) following thoracic SCI contusion. This assessment included the analysis of pro- and anti-inflammatory markers, as well as the expression of critical immunomodulatory kinases, including P38 MAPK, and transcription factors, such as NFκB, which play pivotal roles in injury-induced inflammation. We demonstrate that activated microglia from the injured spinal cord of female rats exhibited a significantly diminutive pro-inflammatory response, but enhanced anti-inflammatory activity compared to males. These changes included lower levels of iNOS and TLR4 expression but increased levels of ARG-1 and CD68 in females after SCI. The altered expression of these markers is indicative of a disparate secretome between the microglia of males and females after SCI and that the female microglia possesses higher phagocytic capabilities (increased CD68). The examination of immunoregulatory kinases and transcription factors revealed that female microglia had higher levels of phosphorylated P38Thr180/Tyr182 MAPK and nuclear NFκB pp50Ser337 but lower amounts of nuclear NFκB pp65Ser536, suggestive of an attenuated pro-inflammatory phenotype in females compared to males after SCI. Collectively, this work provides novel insight into some of the sex disparities that exist in the innate immune response after SCI and indicates that sex is an important variable when designing and testing new therapeutic interventions or interpretating positive or negative responses to an intervention.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Jinyoung Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Ashley N. Burke
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Thomas A. Strong
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Jacqueline Sagen
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
10
|
Pan G, Cui B, Han M, Lin L, Li Y, Wang L, Guo S, Yin Y, Zhan H, Li P. Puerarin inhibits NHE1 activity by interfering with the p38 pathway and attenuates mitochondrial damage induced by myocardial calcium overload in heart failure rats. Acta Biochim Biophys Sin (Shanghai) 2024; 56:270-279. [PMID: 38282474 PMCID: PMC10984851 DOI: 10.3724/abbs.2023269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/11/2023] [Indexed: 01/30/2024] Open
Abstract
Previous studies have shown that puerarin plays a key role in protecting humans and animals from cardiovascular diseases. The exact mechanism of the therapeutic effect of puerarin on various cardiovascular diseases (protective effect on cardiomyocytes) is still unclear. In the present study, we identify the role of puerarin in an animal model of experimental heart failure (HF) and explore its underlying mechanisms. The HF rat model is induced by intraperitoneal injection of adriamycin (ADR), and puerarin is administered intragastrically at low, medium, and high concentrations. We demonstrate that puerarin significantly improves myocardial fibrosis and inflammatory infiltration and, as a result, improves cardiac function in ADR-induced HF rats. Mechanistically, we find for the first time that puerarin inhibits overactivated Na +/H + exchange isoform 1 (NHE1) in HF, which may improve HF by decreasing Na + and Ca 2+ ion concentrations and attenuating mitochondrial damage caused by calcium overload; on the other hand, puerarin inhibits the activation of the p38 pathway in HF, reduces the expressions of TGF-β and proinflammatory cytokines, and suppresses myocardial fibrosis. In conclusion, our results suggest that Puerarin is an effective drug against HF and may play a protective role in the myocardium by inhibiting the activation of p38 and its downstream NHE1.
Collapse
Affiliation(s)
- Guopin Pan
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Baoyue Cui
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
- Nanyang Second General HospitalNanyang473001China
| | - Mingming Han
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Laibiao Lin
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Yinlan Li
- College of PharmacyHeilongjiang University of Chinese MedicineHarbin150040China
| | - Ling Wang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and AngiopathyHubei University of Science and TechnologyXianning437100China
| | - Yaling Yin
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Heqin Zhan
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
| | - Peng Li
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and NeurobiologyHenan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionSchool of Basic Medical SciencesCollege of PharmacyXinxiang Medical UniversityXinxiang453003China
- Hubei Key Laboratory of Diabetes and AngiopathyHubei University of Science and TechnologyXianning437100China
| |
Collapse
|
11
|
Ng GYQ, Loh ZWL, Fann DY, Mallilankaraman K, Arumugam TV, Hande MP. Role of Mitogen-Activated Protein (MAP) Kinase Pathways in Metabolic Diseases. Genome Integr 2024; 15:e20230003. [PMID: 38770527 PMCID: PMC11102075 DOI: 10.14293/genint.14.1.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Physiological processes that govern the normal functioning of mammalian cells are regulated by a myriad of signalling pathways. Mammalian mitogen-activated protein (MAP) kinases constitute one of the major signalling arms and have been broadly classified into four groups that include extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), p38, and ERK5. Each signalling cascade is governed by a wide array of external and cellular stimuli, which play a critical part in mammalian cells in the regulation of various key responses, such as mitogenic growth, differentiation, stress responses, as well as inflammation. This evolutionarily conserved MAP kinase signalling arm is also important for metabolic maintenance, which is tightly coordinated via complicated mechanisms that include the intricate interaction of scaffold proteins, recognition through cognate motifs, action of phosphatases, distinct subcellular localisation, and even post-translational modifications. Aberration in the signalling pathway itself or their regulation has been implicated in the disruption of metabolic homeostasis, which provides a pathophysiological foundation in the development of metabolic syndrome. Metabolic syndrome is an umbrella term that usually includes a group of closely associated metabolic diseases such as hyperglycaemia, hyperlipidaemia, and hypertension. These risk factors exacerbate the development of obesity, diabetes, atherosclerosis, cardiovascular diseases, and hepatic diseases, which have accounted for an increase in the worldwide morbidity and mortality rate. This review aims to summarise recent findings that have implicated MAP kinase signalling in the development of metabolic diseases, highlighting the potential therapeutic targets of this pathway to be investigated further for the attenuation of these diseases.
Collapse
Affiliation(s)
- Gavin Yong Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zachary Wai-Loon Loh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - M. Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
12
|
Zheng Y, Wu T, Hou X, Yang H, Yang Y, Xiu W, Pan Y, Ma Y, Mahemuti A, Xie X. Serum a-1 antitrypsin as a novel biomarker in chronic heart failure. ESC Heart Fail 2023; 10:2865-2874. [PMID: 37417425 PMCID: PMC10567649 DOI: 10.1002/ehf2.14451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/11/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
AIMS Chronic heart failure (CHF) remains a major health issue worldwide. In the present study, we aimed to identify novel circulating biomarkers for CHF using serum proteomics technology and to validate the biomarker in three independent cohorts. METHODS AND RESULTS The isobaric tags for relative and absolute quantitation technology was utilized to identify the potential biomarkers of CHF. The validation was conducted in three independent cohort. Cohort A included 223 patients with ischaemic heart disease (IHD) and 321 patients with ischaemic heart failure (IHF) from the CORFCHD-PCI study. Cohort B recruited 817 patients with IHD and 1139 patients with IHF from the PRACTICE study. Cohort C enrolled 559 non-ischaemic heart disease patients with CHF (n = 316) or without CHF (n = 243). We found the expression of a-1 antitrypsin (AAT) was elevated significantly in patients with CHF compared with that in the patients with stable IHD using statistical and bioinformatics analyses. In a validation study, there was a significant difference between patients with stable IHD and patients with IHF in AAT concentration either in cohort A (1.35 ± 0.40 vs. 1.64 ± 0.56, P < 0.001) or in cohort B (1.37 ± 0.42 vs. 1.70 ± 0.48, P < 0.001). The area under the receiver operating characteristic curve was 0.70 [95% confidence interval (CI): 0.66 to 0.74, P < 0.001] in cohort A and 0.74 (95% CI: 0.72 to 0.76, P < 0.001) in cohort B. Furthermore, AAT was negative correlated with left ventricular ejection fraction (r = -0.261, P < 0.001). After adjusting for confounders using a multivariate logistic regression analysis, AAT remained an independent association with CHF in both cohort A (OR = 3.14, 95% CI: 1.667 to 5.90, P < 0.001) and cohort B (OR = 4.10, 95% CI: 2.97 to 5.65, P < 0.001). This association was also validated in cohort C (OR = 1.86, 95% CI: 1.02 to 3.38, P = 0.043). CONCLUSIONS The present study suggests that serum AAT is a reliable biomarker for CHF in a Chinese population.
Collapse
Affiliation(s)
- Ying‐Ying Zheng
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Ting‐Ting Wu
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Xian‐Geng Hou
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Hai‐Tao Yang
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Yi Yang
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Wen‐Juan Xiu
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Ying Pan
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Yi‐Tong Ma
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Ailiman Mahemuti
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| | - Xiang Xie
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical UniversityNo. 137, Liyushan RoadUrumqi830011XinjiangChina
| |
Collapse
|
13
|
Hong S, Huang W, Zhu X, Tang H, Krier JD, Xing L, Lu B, Gandhi D, Jordan KL, Saadiq IM, Lerman A, Eirin A, Lerman LO. Obesity blunts amelioration of cardiac hypertrophy and fibrosis by human mesenchymal stem/stromal cell-derived extracellular vesicles. Am J Physiol Heart Circ Physiol 2023; 325:H163-H171. [PMID: 37294895 PMCID: PMC10312317 DOI: 10.1152/ajpheart.00676.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/12/2023] [Accepted: 06/01/2023] [Indexed: 06/11/2023]
Abstract
Renovascular hypertension (RVH) can induce cardiac damage that is reversible using adipose tissue-derived mesenchymal stromal/stem cells (A-MSCs). However, A-MSCs isolated from patients with obesity are less effective than lean-A-MSC in blunting hypertensive cardiomyopathy in mice with RVH. We tested the hypothesis that this impairment extends to their obese A-MSC-extracellular vesicles (EVs) progeny. MSCs were harvested from the subcutaneous fat of obese and lean human subjects, and their EVs were collected and injected into the aorta of mice 2 wk after renal artery stenosis or sham surgery. Cardiac left ventricular (LV) function was studied with MRI 2 wk later, and myocardial tissue ex vivo. Blood pressure, LV myocardial wall thickness, mass, and fibrosis that were elevated in RVH mice were suppressed only by lean EVs. Hence, human A-MSC-derived lean EVs are more effective than obese EVs in blunting hypertensive cardiac injury in RVH mice. These observations highlight impaired paracrine repair potency of endogenous MSCs in patients with obesity.NEW & NOTEWORTHY Injection of A-MSC-derived EVs harvested from patients who are lean can resolve myocardial injury in mice with experimental renovascular hypertension more effectively than A-MSC-derived EVs from patients with obesity. These observations underscore and might have important ramifications for the self-healing capacity of patients with obesity and for the use of autologous EVs as a regenerative tool.
Collapse
Affiliation(s)
- Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Li Xing
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Bo Lu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Deep Gandhi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
14
|
Papanicolaou KN, Jung J, Ashok D, Zhang W, Modaressanavi A, Avila E, Foster DB, Zachara NE, O'Rourke B. Inhibiting O-GlcNAcylation impacts p38 and Erk1/2 signaling and perturbs cardiomyocyte hypertrophy. J Biol Chem 2023; 299:102907. [PMID: 36642184 PMCID: PMC9988579 DOI: 10.1016/j.jbc.2023.102907] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The dynamic cycling of O-linked GlcNAc (O-GlcNAc) on and off Ser/Thr residues of intracellular proteins, termed O-GlcNAcylation, is mediated by the conserved enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase. O-GlcNAc cycling is important in homeostatic and stress responses, and its perturbation sensitizes the heart to ischemic and other injuries. Despite considerable progress, many molecular pathways impacted by O-GlcNAcylation in the heart remain unclear. The mitogen-activated protein kinase (MAPK) pathway is a central signaling cascade that coordinates developmental, physiological, and pathological responses in the heart. The developmental or adaptive arm of MAPK signaling is primarily mediated by Erk kinases, while the pathophysiologic arm is mediated by p38 and Jnk kinases. Here, we examine whether O-GlcNAcylation affects MAPK signaling in cardiac myocytes, focusing on Erk1/2 and p38 in basal and hypertrophic conditions induced by phenylephrine. Using metabolic labeling of glycans coupled with alkyne-azide "click" chemistry, we found that Erk1/2 and p38 are O-GlcNAcylated. Supporting the regulation of p38 by O-GlcNAcylation, the OGT inhibitor, OSMI-1, triggers the phosphorylation of p38, an event that involves the NOX2-Ask1-MKK3/6 signaling axis and also the noncanonical activator Tab1. Additionally, OGT inhibition blocks the phenylephrine-induced phosphorylation of Erk1/2. Consistent with perturbed MAPK signaling, OSMI-1-treated cardiomyocytes have a blunted hypertrophic response to phenylephrine, decreased expression of cTnT (key component of the contractile apparatus), and increased expression of maladaptive natriuretic factors Anp and Bnp. Collectively, these studies highlight new roles for O-GlcNAcylation in maintaining a balanced activity of Erk1/2 and p38 MAPKs during hypertrophic growth responses in cardiomyocytes.
Collapse
Affiliation(s)
- Kyriakos N Papanicolaou
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Jessica Jung
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Deepthi Ashok
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wenxi Zhang
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Modaressanavi
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eddie Avila
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natasha E Zachara
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
A comparison of electrosprayed vs vortexed glycosaminoglycan-peptide nanoparticle platform for protection and improved delivery of therapeutic peptides. Colloids Surf B Biointerfaces 2023; 222:113112. [PMID: 36599186 DOI: 10.1016/j.colsurfb.2022.113112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Therapeutic peptides capable of reducing inflammation via inhibition of the MAP kinase 2 pathway have the potential to reduce inflammation in atopic dermatitis by suppressing secretion of inflammatory cytokines by resident keratinocytes. One of the biggest hurdles to the use of therapeutic peptides, however, is their rapid degradation by intrinsic proteases and peptidases found in serum. Here we introduce a new nanoparticle technology that enhances and extends the bioactivity of a MAP KAP kinase 2 inhibitor peptide (MK2i) via electrostatic complexation with Dermatan sulfate (DS), a glycosaminoglycan, and explore their properties under various conditions. DS-MK2i nanoparticles can be made using electrospray ionization or sonication and vortexing with no stabilizing polymers or crosslinking. Average particle diameter, polydispersity index, and zeta potential were measured over a pH range of 2.5-11.5, in increments of 0.5, in water and at physiological ionic strength. Both particle types were shown to be shelf stable, robust, and behave differently in response to pH. They are also significantly more effective at suppressing cytokine secretion in inflamed, human keratinocytes than peptide alone in the presence of serum, providing a facile method of protecting peptides for therapeutic delivery in conditions such as atopic dermatitis, and abrogating the need for serum-starvation in in vitro testing.
Collapse
|
16
|
Immunoproteasome Inhibition Ameliorates Aged Dystrophic Mouse Muscle Environment. Int J Mol Sci 2022; 23:ijms232314657. [PMID: 36498987 PMCID: PMC9739773 DOI: 10.3390/ijms232314657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Muscle wasting is a major pathological feature observed in Duchenne muscular dystrophy (DMD) and is the result of the concerted effects of inflammation, oxidative stress and cell senescence. The inducible form of proteasome, or immunoproteasome (IP), is involved in all the above mentioned processes, regulating antigen presentation, cytokine production and immune cell response. IP inhibition has been previously shown to dampen the altered molecular, histological and functional features of 3-month-old mdx mice, the animal model for DMD. In this study, we described the role of ONX-0914, a selective inhibitor of the PSMB8 subunit of immunoproteasome, in ameliorating the pathological traits that could promote muscle wasting progression in older, 9-month-old mdx mice. ONX-0914 reduces the number of macrophages and effector memory T cells in muscle and spleen, while increasing the number of regulatory T cells. It modulates inflammatory markers both in skeletal and cardiac muscle, possibly counteracting heart remodeling and hypertrophy. Moreover, it buffers oxidative stress by improving mitochondrial efficiency. These changes ultimately lead to a marked decrease of fibrosis and, potentially, to more controlled myofiber degeneration/regeneration cycles. Therefore, ONX-0914 is a promising molecule that may slow down muscle mass loss, with relatively low side effects, in dystrophic patients with moderate to advanced disease.
Collapse
|
17
|
Koniari I, Artopoulou E, Velissaris D, Ainslie M, Mplani V, Karavasili G, Kounis N, Tsigkas G. Biomarkers in the clinical management of patients with atrial fibrillation and heart failure. J Geriatr Cardiol 2021; 18:908-951. [PMID: 34908928 PMCID: PMC8648548 DOI: 10.11909/j.issn.1671-5411.2021.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) are two cardiovascular diseases with an increasing prevalence worldwide. These conditions share common pathophysiologiesand frequently co-exit. In fact, the occurrence of either condition can 'cause' the development of the other, creating a new patient group that demands different management strategies to that if they occur in isolation. Regardless of the temproral association of the two conditions, their presence is linked with adverse cardiovascular outcomes, increased rate of hospitalizations, and increased economic burden on healthcare systems. The use of low-cost, easily accessible and applicable biomarkers may hasten the correct diagnosis and the effective treatment of AF and HF. Both AF and HF effect multiple physiological pathways and thus a great number of biomarkers can be measured that potentially give the clinician important diagnostic and prognostic information. These will then guide patient centred therapeutic management. The current biomarkers that offer potential for guiding therapy, focus on the physiological pathways of miRNA, myocardial stretch and injury, oxidative stress, inflammation, fibrosis, coagulation and renal impairment. Each of these has different utility in current clinincal practice.
Collapse
Affiliation(s)
- Ioanna Koniari
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | | | - Mark Ainslie
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
- Division of Cardiovascular Sciences, University of Manchester
| | - Virginia Mplani
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Georgia Karavasili
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Nicholas Kounis
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| |
Collapse
|
18
|
Ma SQ, Guo Z, Liu FY, Hasan SG, Yang D, Tang N, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. 6-Gingerol protects against cardiac remodeling by inhibiting the p38 mitogen-activated protein kinase pathway. Acta Pharmacol Sin 2021; 42:1575-1586. [PMID: 33462378 PMCID: PMC8463710 DOI: 10.1038/s41401-020-00587-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/20/2020] [Indexed: 02/02/2023]
Abstract
6-Gingerol, a pungent ingredient of ginger, has been reported to possess anti-inflammatory and antioxidant activities, but the effect of 6-gingerol on pressure overload-induced cardiac remodeling remains inconclusive. In this study, we investigated the effect of 6-gingerol on cardiac remodeling in in vivo and in vitro models, and to clarify the underlying mechanisms. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with 6-gingerol (20 mg/kg, ig) three times a week (1 week in advance and continued until the end of the experiment). Four weeks after TAC surgery, the mice were subjected to echocardiography, and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes and cardiac fibroblasts were used to validate the protective effects of 6-gingerol in response to phenylephrine (PE) and transforming growth factor-β (TGF-β) challenge. We showed that 6-gingerol administration protected against pressure overload-induced cardiac hypertrophy, fibrosis, inflammation, and dysfunction in TAC mice. In the in vitro study, we showed that treatment with 6-gingerol (20 μM) blocked PE-induced-cardiomyocyte hypertrophy and TGF-β-induced cardiac fibroblast activation. Furthermore, 6-gingerol treatment significantly decreased mitogen-activated protein kinase p38 (p38) phosphorylation in response to pressure overload in vivo and extracellular stimuli in vitro, which was upregulated in the absence of 6-gingerol treatment. Moreover, transfection with mitogen-activated protein kinase kinase 6 expressing adenoviruses (Ad-MKK6), which specifically activated p38, abolished the protective effects of 6-gingerol in both in vitro and in vivo models. In conclusion, 6-gingerol improves cardiac function and alleviates cardiac remodeling induced by pressure overload in a p38-dependent manner. The present study demonstrates that 6-gingerol is a promising agent for the intervention of pathological cardiac remodeling.
Collapse
Affiliation(s)
- Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Shahzad-Gul Hasan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
- Department of Medicine, Bahawal Victoria Hospital, Bahawalpur, 63100, Pakistan
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| |
Collapse
|
19
|
Chen X, Yun C, Zheng H, Chen X, Han Q, Pan H, Wang Y, Zhong J. The protective effects of S14G-humanin (HNG) against streptozotocin (STZ)-induced cardiac dysfunction. Bioengineered 2021; 12:5491-5503. [PMID: 34506248 PMCID: PMC8806847 DOI: 10.1080/21655979.2021.1964894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Excessive oxidative stress, inflammation, and myocardial hypertrophy have been associated with diabetic cardiomyopathy (DCM). S14G-humanin (HNG) is a potent humanin analogue that has demonstrated cytoprotective effects in a variety of cells and tissues. However, the pharmacological function of HNG in diabetic cardiomyopathy has not yet been reported. In the present study, we investigated the protective effects of HNG against streptozotocin (STZ)-induced cardiac dysfunction in diabetic mice. Myocardial hypertrophy in diabetic mice was determined using Wheat Gem Agglutinin (WGA) staining. The heart function was measured with Echocardiographic imaging. Levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) proteins in plasma were measured using enzyme-linked immunosorbent assay (ELISA) kits. Protein expression of Phosphorylated p38/p38 was determined using western blots. We found that HNG treatment attenuated the STZ-induced myocardial hypertrophy and significantly improved heart function. Also, its treatment proved effective as it reduced the levels of several myocardial injury indicators, including creatine kinase-MB (CK-MB), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and both the cardiac and plasma levels of TNF-α and IL-6, highlighting its effect on the STZ-induced myocardial injury. Lastly, HNG suppressed the activation of the p38/nuclear factor kappa-B (NF-κB) signaling pathway. S14G humanin possesses protective effects against streptozotocin-induced cardiac dysfunction through inhibiting the activation of the p38/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaopan Chen
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Chuan Yun
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Hailong Zheng
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Xu Chen
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Qianfei Han
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Hua Pan
- Department Of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Yang Wang
- Department Of Cardiology, Affiliated Haikou Hospital Of Xiangya Medical College, Central South University, Haikou, Hainan Province, China
| | - Jianghua Zhong
- Department Of Cardiology, Affiliated Haikou Hospital Of Xiangya Medical College, Central South University, Haikou, Hainan Province, China
| |
Collapse
|
20
|
Eirin A, Meng Y, Zhu XY, Li Y, Saadiq IM, Jordan KL, Tang H, Lerman A, van Wijnen AJ, Lerman LO. The Micro-RNA Cargo of Extracellular Vesicles Released by Human Adipose Tissue-Derived Mesenchymal Stem Cells Is Modified by Obesity. Front Cell Dev Biol 2021; 9:660851. [PMID: 34095124 PMCID: PMC8173369 DOI: 10.3389/fcell.2021.660851] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
Obesity is a chronic disease that interferes with normal repair processes, including adipose mesenchymal stem/stromal cells (ASCs) function. ASCs produce extracellular vesicles (EVs) that activate a repair program in recipient cells partly via their micro-RNA (miRNA) cargo. We hypothesized that obesity alters the miRNA expression profile of human ASC-derived EVs, limiting their capacity to repair injured cells. Human ASCs were harvested from obese and age- and gender-matched non-obese (lean) subjects during bariatric or cosmetic surgeries, respectively (n = 5 each), and their EVs isolated. Following high-throughput sequencing analysis, differentially expressed miRNAs were identified and their gene targets classified based on cellular component, molecular function, and biological process. The capacity of human lean- and obese-EVs to modulate inflammation, apoptosis, as well as mitogen-activated protein kinase (MAPK) and Wnt signaling in injured human proximal tubular epithelial (HK2) cells was evaluated in vitro. The number of EVs released from lean- and obese-ASCs was similar, but obese-EVs were smaller compared to lean-EVs. Differential expression analysis revealed 8 miRNAs upregulated (fold change > 1.4, p < 0.05) and 75 downregulated (fold change < 0.7, p < 0.05) in obese-EVs vs. lean-EVs. miRNAs upregulated in obese-EVs participate in regulation of NFk-B and MAPK signaling, cytoskeleton organization, and apoptosis, whereas those downregulated in obese-EVs are implicated in cell cycle, angiogenesis, and Wnt and MAPK signaling. Treatment of injured HK2 cells with obese-EVs failed to decrease inflammation, and they decreased apoptosis and MAPK signaling significantly less effectively than their lean counterparts. Obesity alters the size and miRNA cargo of human ASC-derived EVs, as well as their ability to modulate important injury pathways in recipient cells. These observations may guide development of novel strategies to improve healing and repair in obese individuals.
Collapse
Affiliation(s)
- Alfonso Eirin
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Yu Meng
- Department of Nephrology, The First Hospital Affiliated to Jinan University, Guangzhou, China
| | - Xiang-Yang Zhu
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Yongxin Li
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Ishran M. Saadiq
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Kyra L. Jordan
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Hui Tang
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - Lilach O. Lerman
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
21
|
Mimpen JY, Baldwin MJ, Cribbs AP, Philpott M, Carr AJ, Dakin SG, Snelling SJB. Interleukin-17A Causes Osteoarthritis-Like Transcriptional Changes in Human Osteoarthritis-Derived Chondrocytes and Synovial Fibroblasts In Vitro. Front Immunol 2021; 12:676173. [PMID: 34054865 PMCID: PMC8153485 DOI: 10.3389/fimmu.2021.676173] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
Increased interleukin (IL)-17A has been identified in joints affected by osteoarthritis (OA), but it is unclear how IL-17A, and its family members IL-17AF and IL-17F, can contribute to human OA pathophysiology. Therefore, we aimed to evaluate the gene expression and signalling pathway activation effects of the different IL-17 family members in chondrocytes and synovial fibroblasts derived from cartilage and synovium of patients with end-stage knee OA. Immunohistochemistry staining confirmed that IL-17 receptor A (IL-17RA) and IL-17RC are expressed in end-stage OA-derived cartilage and synovium. Chondrocytes and synovial fibroblasts derived from end-stage OA patients were treated with IL-17A, IL-17AF, or IL-17F, and gene expression was assessed with bulk RNA-Seq. Hallmark pathway analysis showed that IL-17 cytokines regulated several OA pathophysiology-related pathways including immune-, angiogenesis-, and complement-pathways in both chondrocytes and synovial fibroblasts derived from end-stage OA patients. While overall IL-17A induced the strongest transcriptional response, followed by IL-17AF and IL-17F, not all genes followed this pattern. Disease-Gene Network analysis revealed that IL-17A-related changes in gene expression in these cells are associated with experimental arthritis, knee arthritis, and musculoskeletal disease gene-sets. Western blot analysis confirmed that IL-17A significantly activates p38 and p65 NF-κB. Incubation of chondrocytes and synovial fibroblasts with anti-IL-17A monoclonal antibody secukinumab significantly inhibited IL-17A-induced gene expression. In conclusion, the association of IL-17-induced transcriptional changes with arthritic gene-sets supports a role for IL-17A in OA pathophysiology. Future studies should further investigate the role of IL-17A in the OA joint to establish whether anti-IL-17 treatment could be a potential therapeutic option in OA patients with an inflammatory phenotype.
Collapse
Affiliation(s)
- Jolet Y Mimpen
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Mathew J Baldwin
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Adam P Cribbs
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Martin Philpott
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Andrew J Carr
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Stephanie G Dakin
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Sarah J B Snelling
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Wang Z, Zeng Y, Lin H. The effect of chronic intermittent hypoxia on atherosclerosis in rat offspring. Int J Cardiol 2021; 335:98-103. [PMID: 33957177 DOI: 10.1016/j.ijcard.2021.04.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/19/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND To determine whether rat offspring born under conditions of chronic intermittent hypoxia (CIH) would develop systemic inflammation and be susceptible to CIH-mediated injury on artery. METHODS CIH rat model was established. The expression levels of p38MAPK, NF-κB p65, CRP, TNFα, and IL-8 were measured. The arterial pathology of offspring whose mothers were exposed to CIH during pregnancy was evaluated. RESULTS The levels of p-P38MAPK and NF-κB p65 were significantly up-regulated following induction of intra-uterine CIH Induction of intra- and extra-uterine CIH had an interaction regarding the expression profiles of these markers (P < 0.01, P < 0.01, P < 0.01, P < 0.01, P = 0.020, respectively). Pathologic analysis of the arteries confirmed that intra-uterine CIH increased the tunica intima thickness (P < 0.01), but had no effect on the tunica media (P = 0.974). Furthermore, induction of intra- and extra-uterine CIH had a synergistic interaction on tunica intima thickness (P = 0.004). CONCLUSIONS Intra-uterine CIH caused tunica intima thickening and development of pre-atherosclerotic lesions in offspring via NF-κB p65 and p-p38 MAPK. Furthermore, the expression of NF-κB p65 and p-p38 MAPK and the extent of pre-atherosclerotic lesions were either exacerbated or alleviated in offspring when re-exposed to CIH later.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Huihuang Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
23
|
Prasher P, Sharma M, Zacconi F, Gupta G, Aljabali AA, Mishra V, Tambuwala MM, Kapoor DN, Negi P, Andreoli Pinto TDJ, Singh I, Chellappan DK, Dua K. Synthesis and Anticancer Properties of ‘Azole’ Based Chemotherapeutics as Emerging Chemical Moieties: A Comprehensive Review. CURR ORG CHEM 2021. [DOI: 10.2174/1385272824999200820152501] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Azole frameworks serve as privileged scaffolds in the contemporary drug design
paradigm owing to their unique physicochemical profile that promotes the development
of highly selective, physiological benevolent chemotherapeutics. Several azole nuclei
function as bioisostere in medicinal chemistry and prompt the development of tailored
therapeutics for targeting the desired biological entities. Besides, the azole scaffold forms
an integral part in the advanced drug designing methodologies, such as target template insitu
drug synthesis, that assists in rapid identification of the hit molecules form a diverse
pool of leads; and direct biomolecule-drug conjugation, along with bioorthogonal strategies
that ensure localization, and superior target specificity of the directed therapeutic.
Lastly, the structural diversity of azole framework and high yielding click synthetic methods
provide a comprehensive Structure-Activity Relationship analysis for design optimization of the potential
drug molecules by fine-tuning the placement of different substituents critical for the activity. This review provides
a comprehensive analysis of the synthesis and anticancer potential of azole based chemotherapeutics.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Arcadia Grant, Dehradun 248007, India
| | - Flavia Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago 7820436, Chile
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302 017, Jaipur, India
| | - Alaa A.A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Deepak N. Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Terezinha de Jesus Andreoli Pinto
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, Professor Lineu Prestes Street, São Paulo 05508-000, Brazil
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
24
|
Choi H, Dey AK, Sharma G, Bhoite R, Burkholder G, Fedson S, Jneid H. Etiology and pathophysiology of heart failure in people with HIV. Heart Fail Rev 2021; 26:497-505. [PMID: 33619685 DOI: 10.1007/s10741-020-10048-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 11/25/2022]
Abstract
HIV-associated cardiomyopathy is a well-established sequela in people infected with HIV (PHIV). Despite significant advances in HIV management through the use of highly active anti-retroviral therapy (HAART), PHIV on HAART continue to have elevated risk of cardiomyopathy and heart failure, even when accounting for known cardiovascular risk factors. This review article will explore the proposed mechanisms by which chronic HIV infection induces cardiomyopathy and heart failure in the setting of HAART. Evaluation, work-up, and management of cardiomyopathy in PHIV will also be briefly discussed. The advent of HAART has altered the pathophysiology HIV-associated cardiomyopathy from a rapidly progressive cardiomyopathy, often with pericardial involvement, into a chronic process involving inflammation and persistent immune dysregulation. With the significant decrease in AIDS-related deaths, the prevalence of cardiomyopathy and the mortality associated with heart failure in PHIV have increased. Multiple immune-related and inflammatory mechanisms have been proposed, which may provide insight into evaluation and management of cardiomyopathy in PHIV.
Collapse
Affiliation(s)
- Harry Choi
- Section of Inflammation and Cardiometabolic Diseases, National Institute of Health, Bethesda, MD, USA
| | - Amit K Dey
- Section of Inflammation and Cardiometabolic Diseases, National Institute of Health, Bethesda, MD, USA
| | - Gaurav Sharma
- Division of Medicine, University of South Alabama, Mobile, AL, USA
| | - Rahul Bhoite
- Division of Medicine, MedStar Union Memorial Hospital and Good Samaritan Hospital, Baltimore, MD, USA
| | - Greer Burkholder
- Division of Infectious Diseases, University of Alabama, Birmingham, AL, USA
| | - Savitri Fedson
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VA Hospital, Houston, TX, 77030, USA
| | - Hani Jneid
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VA Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Chiang MT, Chen IM, Hsu FF, Chen YH, Tsai MS, Hsu YW, Leu HB, Huang PH, Chen JW, Liu FT, Chen YH, Chau LY. Gal-1 (Galectin-1) Upregulation Contributes to Abdominal Aortic Aneurysm Progression by Enhancing Vascular Inflammation. Arterioscler Thromb Vasc Biol 2021; 41:331-345. [PMID: 33147994 DOI: 10.1161/atvbaha.120.315398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a vascular degenerative disease causing sudden rupture of aorta and significant mortality in elders. Nevertheless, no prognostic and therapeutic target is available for disease management. Gal-1 (galectin-1) is a β-galactoside-binding lectin constitutively expressed in vasculature with roles in maintaining vascular homeostasis. This study aims to investigate the potential involvement of Gal-1 in AAA progression. Approach and Results: Gal-1 was significantly elevated in circulation and aortic tissues of Ang II (angiotensin II)-infused apoE-deficient mice developing AAA. Gal-1 deficiency reduced incidence and severity of AAA with lower expression of aortic MMPs (matrix metalloproteases) and proinflammatory cytokines. TNFα (tumor necrosis factor alpha) induced Gal-1 expression in cultured vascular smooth muscle cells and adventitial fibroblasts. Gal-1 deletion enhanced TNFα-induced MMP9 expression in fibroblasts but not vascular smooth muscle cells. Cysteinyl-labeling assay demonstrated that aortic Gal-1 exhibited susceptibility to oxidation in vivo. Recombinant oxidized Gal-1 induced expression of MMP9 and inflammatory cytokines to various extents in macrophages, vascular smooth muscle cells, and fibroblasts through activation of MAP (mitogen-activated protein) kinase signaling. Clinically, serum MMP9 level was significantly higher in both patients with AAA and coronary artery disease than in control subjects, whereas serum Gal-1 level was elevated in patients with AAA but not coronary artery disease when compared with controls. CONCLUSIONS Gal-1 is highly induced and contributes to AAA by enhancing matrix degradation activity and inflammatory responses in experimental model. The pathological link between Gal-1 and AAA is also observed in human patients. These findings support the potential of Gal-1 as a disease biomarker and therapeutic target of AAA.
Collapse
MESH Headings
- Adventitia/metabolism
- Adventitia/pathology
- Angiotensin II
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortitis/chemically induced
- Aortitis/metabolism
- Aortitis/pathology
- Case-Control Studies
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Disease Progression
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Galectin 1/blood
- Galectin 1/deficiency
- Galectin 1/genetics
- Galectin 1/metabolism
- Humans
- Inflammation Mediators/metabolism
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Male
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Signal Transduction
- Up-Regulation
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Ming-Tsai Chiang
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - I-Ming Chen
- Division of Cardiovascular Surgery, Department of Surgery (I.-M.C.), Taipei Veterans General Hospital, Taiwan
| | - Fu-Fei Hsu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Yen-Hui Chen
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Min-Shao Tsai
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Yaw-Wen Hsu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Hsin-Bang Leu
- Division of Healthcare and Management, Healthcare Center (H.-B.L.), Taipei Veterans General Hospital, Taiwan
- Department of Medicine, School of Medicine (H.-B.L., Ying-Hwa Chen), National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Institute of Clinical Medicine (P.-H.H.), National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Institute of Pharmacology (J.-W.C.), National Yang-Ming University, Taipei, Taiwan
| | - Fu-Tong Liu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Department of Medicine, School of Medicine (H.-B.L., Ying-Hwa Chen), National Yang-Ming University, Taipei, Taiwan
| | - Lee-Young Chau
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| |
Collapse
|
26
|
p38 MAPK Pathway in the Heart: New Insights in Health and Disease. Int J Mol Sci 2020; 21:ijms21197412. [PMID: 33049962 PMCID: PMC7582802 DOI: 10.3390/ijms21197412] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The p38 mitogen-activated kinase (MAPK) family controls cell adaptation to stress stimuli. p38 function has been studied in depth in relation to cardiac development and function. The first isoform demonstrated to play an important role in cardiac development was p38α; however, all p38 family members are now known to collaborate in different aspects of cardiomyocyte differentiation and growth. p38 family members have been proposed to have protective and deleterious actions in the stressed myocardium, with the outcome of their action in part dependent on the model system under study and the identity of the activated p38 family member. Most studies to date have been performed with inhibitors that are not isoform-specific, and, consequently, knowledge remains very limited about how the different p38s control cardiac physiology and respond to cardiac stress. In this review, we summarize the current understanding of the role of the p38 pathway in cardiac physiology and discuss recent advances in the field.
Collapse
|
27
|
|
28
|
Lubrano V, Vergaro G, Maltinti M, Ghionzoli N, Emdin M, Papa A. α-1 Antitrypsin as a potential biomarker in chronic heart failure. J Cardiovasc Med (Hagerstown) 2020; 21:209-215. [DOI: 10.2459/jcm.0000000000000937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
29
|
Oldfield CJ, Duhamel TA, Dhalla NS. Mechanisms for the transition from physiological to pathological cardiac hypertrophy. Can J Physiol Pharmacol 2020; 98:74-84. [DOI: 10.1139/cjpp-2019-0566] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The heart is capable of responding to stressful situations by increasing muscle mass, which is broadly defined as cardiac hypertrophy. This phenomenon minimizes ventricular wall stress for the heart undergoing a greater than normal workload. At initial stages, cardiac hypertrophy is associated with normal or enhanced cardiac function and is considered to be adaptive or physiological; however, at later stages, if the stimulus is not removed, it is associated with contractile dysfunction and is termed as pathological cardiac hypertrophy. It is during physiological cardiac hypertrophy where the function of subcellular organelles, including the sarcolemma, sarcoplasmic reticulum, mitochondria, and myofibrils, may be upregulated, while pathological cardiac hypertrophy is associated with downregulation of these subcellular activities. The transition of physiological cardiac hypertrophy to pathological cardiac hypertrophy may be due to the reduction in blood supply to hypertrophied myocardium as a consequence of reduced capillary density. Oxidative stress, inflammatory processes, Ca2+-handling abnormalities, and apoptosis in cardiomyocytes are suggested to play a critical role in the depression of contractile function during the development of pathological hypertrophy.
Collapse
Affiliation(s)
- Christopher J. Oldfield
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Todd A. Duhamel
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
30
|
Role of oxidative stress-related biomarkers in heart failure: galectin 3, α1-antitrypsin and LOX-1: new therapeutic perspective? Mol Cell Biochem 2019; 464:143-152. [DOI: 10.1007/s11010-019-03656-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023]
|
31
|
Roffe-Vazquez DN, Huerta-Delgado AS, Castillo EC, Villarreal-Calderón JR, Gonzalez-Gil AM, Enriquez C, Garcia-Rivas G, Elizondo-Montemayor L. Correlation of Vitamin D with Inflammatory Cytokines, Atherosclerotic Parameters, and Lifestyle Factors in the Setting of Heart Failure: A 12-Month Follow-Up Study. Int J Mol Sci 2019; 20:ijms20225811. [PMID: 31752330 PMCID: PMC6887713 DOI: 10.3390/ijms20225811] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Vitamin D deficiency is highly prevalent worldwide. It has been associated with heart failure (HF) given its immunoregulatory functions. In-vitro and animal models have shown protective roles through mechanisms involving procollagen-1, JNK2, calcineurin/NFAT, NF-κB, MAPK, Th1, Th2, Th17, cytokines, cholesterol-efflux, oxLDL, and GLUT4, among others. A 12-month follow-up in HF patients showed a high prevalence of vitamin D deficiency, with no seasonal variation (64.7-82.4%). A positive correlation between serum 25(OH)D concentration and dietary intake of vitamin D-rich foods was found. A significant inverse correlation with IL-1β (R = -0.78), TNF-α (R = -0.53), IL-6 (R = -0.42), IL-8 (R = -0.41), IL-17A (R = -0.31), LDL-cholesterol (R = -0.51), Apo-B (R = -0.57), total-cholesterol (R = -0.48), and triglycerides (R = -0.32) was shown. Cluster analysis demonstrated that patients from cluster three, with the lowest 25(OH)D levels, presented the lowermost vitamin D intake, IL-10 (1.0 ± 0.9 pg/mL), and IL-12p70 (0.5 ± 0.4 pg/mL), but the highest TNF-α (9.1 ± 3.5 pg/mL), IL-8 (55.6 ± 117.1 pg/mL), IL-17A (3.5 ± 2.0 pg/mL), total-cholesterol (193.9 ± 61.4 mg/dL), LDL-cholesterol (127.7 ± 58.2 mg/dL), and Apo-B (101.4 ± 33.4 mg/dL) levels, compared with patients from cluster one. Although the role of vitamin D in the pathogenesis of HF in humans is still uncertain, we applied the molecular mechanisms of in-vitro and animal models to explain our findings. Vitamin D deficiency might contribute to inflammation, remodeling, fibrosis, and atherosclerosis in patients with HF.
Collapse
Affiliation(s)
- Daniel N. Roffe-Vazquez
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition, Escuela de Medicina, Monterrey 64710, N.L., Mexico; (D.N.R.-V.); (A.S.H.-D.); (J.R.V.-C.); (A.M.G.-G.)
| | - Anna S. Huerta-Delgado
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition, Escuela de Medicina, Monterrey 64710, N.L., Mexico; (D.N.R.-V.); (A.S.H.-D.); (J.R.V.-C.); (A.M.G.-G.)
| | - Elena C. Castillo
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, San Pedro Garza-Garcia 66278, N.L., Mexico; (E.C.C.); (C.E.)
| | - José R. Villarreal-Calderón
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition, Escuela de Medicina, Monterrey 64710, N.L., Mexico; (D.N.R.-V.); (A.S.H.-D.); (J.R.V.-C.); (A.M.G.-G.)
| | - Adrian M. Gonzalez-Gil
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition, Escuela de Medicina, Monterrey 64710, N.L., Mexico; (D.N.R.-V.); (A.S.H.-D.); (J.R.V.-C.); (A.M.G.-G.)
| | - Cecilio Enriquez
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, San Pedro Garza-Garcia 66278, N.L., Mexico; (E.C.C.); (C.E.)
| | - Gerardo Garcia-Rivas
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, San Pedro Garza-Garcia 66278, N.L., Mexico; (E.C.C.); (C.E.)
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Escuela de Medicina, San Pedro Garza-Garcia 66278, N.L., Mexico
- Correspondence: (G.G.-R.); (L.E.-M.)
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition, Escuela de Medicina, Monterrey 64710, N.L., Mexico; (D.N.R.-V.); (A.S.H.-D.); (J.R.V.-C.); (A.M.G.-G.)
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Escuela de Medicina, San Pedro Garza-Garcia 66278, N.L., Mexico
- Correspondence: (G.G.-R.); (L.E.-M.)
| |
Collapse
|
32
|
Analysis of potential roles of combinatorial microRNA regulation in occurrence of valvular heart disease with atrial fibrillation based on computational evidences. PLoS One 2019; 14:e0221900. [PMID: 31479479 PMCID: PMC6719876 DOI: 10.1371/journal.pone.0221900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/16/2019] [Indexed: 11/19/2022] Open
Abstract
Background Atrial fibrillation (AF) is the most common arrhythmia. Patients with valvular heart disease (VHD) frequently have AF. Growing evidence demonstrates that a specifically altered pattern of microRNA (miRNA) expression is related to valvular heart disease with atrial fibrillation (AF-VHD) processes. However, the combinatorial regulation by multiple miRNAs in inducing AF-VHD remains largely unknown. Methods The work identified AF-VHD-specific miRNAs and their combinations through mapping miRNA expression profile into differential co-expression network. The expressions of some dysregulated miRNAs were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The regulations of signaling pathways by the combinatorial miRNAs were predicted by enrichment analysis tools. Results Thirty-two differentially expressed (DE) miRNAs were identified to be AF-VHD-specific, some of which were new findings. These miRNAs interacted to form 5 combinations. qRT-PCR confirmed the different expression of several identified miRNAs, which illustrated the reliability and biomarker potentials of 32 dysregulation miRNAs. The biological characteristics of combinatorial miRNAs related to AF-VHD were highlighted. Twelve signaling pathways regulated by combinatorial miRNAs were predicted to be possibly associated with AF-VHD. Conclusions The AF-VHD-related signaling pathways regulated by combinatorial miRNAs may play an important role in the occurrence of AF-VHD. The work brings new insights into biomarkers and miRNA combination regulation mechanism in AF-VHD as well as further biological experiments.
Collapse
|
33
|
Matsumura N, Zordoky BN, Robertson IM, Hamza SM, Parajuli N, Soltys CLM, Beker DL, Grant MK, Razzoli M, Bartolomucci A, Dyck JRB. Co-administration of resveratrol with doxorubicin in young mice attenuates detrimental late-occurring cardiovascular changes. Cardiovasc Res 2019; 114:1350-1359. [PMID: 29566148 DOI: 10.1093/cvr/cvy064] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/15/2018] [Indexed: 12/23/2022] Open
Abstract
Aims Doxorubicin (DOX) is among the most effective chemotherapies used in paediatric cancer patients. However, the clinical utility of DOX is offset by its well-known cardiotoxicity, which often does not appear until later in life. Since hypertension significantly increases the risk of late-onset heart failure in childhood cancer survivors, we investigated whether juvenile DOX exposure impairs the ability to adapt to angiotensin II (Ang II)-induced hypertension later in life and tested a treatment that could prevent this. Methods and results Five-week-old male mice were administered a low dose of DOX (4 mg/kg) or saline once a week for 3 weeks and then allowed to recover for 5 weeks. Following the 5-week recovery period, mice were infused with Ang II or saline for 2 weeks. In another cohort, mice were fed chow containing 0.4% resveratrol 1 week before, during, and 1 week after the DOX administrations. One week after the last DOX administration, p38 mitogen-activated protein kinase (MAPK) was activated in hearts of DOX-treated mice demonstrating molecular signs of cardiac stress; yet, there was no change in cardiac function between groups. However, DOX-treated mice failed to develop compensatory cardiac hypertrophy in response to Ang II-induced hypertension later in life. Of importance, mice receiving DOX with resveratrol co-administration displayed normalization in p38 MAPK activation in the heart and a restored capacity for cardiac hypertrophy in response to Ang II-induced hypertension. Conclusion We have developed a juvenile mouse model of DOX-induced cardiotoxicity that displays no immediate overt physiological dysfunction; but, leads to an impaired ability of the heart to adapt to hypertension later in life. We also show that co-administration of resveratrol during DOX treatment was sufficient to normalize molecular markers of cardiotoxicity and restore the ability of the heart to undergo adaptive remodelling in response to hypertension later in life.
Collapse
Affiliation(s)
- Nobutoshi Matsumura
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada.,Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Beshay N Zordoky
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 308 Harvard Street S.E., Minneapolis, MN 55455, USA
| | - Ian M Robertson
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| | - Shereen M Hamza
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| | - Nirmal Parajuli
- Department of Medicine, Cardiovascular Research Centre, Faculty of Medicine and Dentistry, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| | - Carrie-Lynn M Soltys
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| | - Donna L Beker
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| | - Marianne K Grant
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 308 Harvard Street S.E., Minneapolis, MN 55455, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, 2231 6th Street S.E. Minneapolis, MN 55455, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, 2231 6th Street S.E. Minneapolis, MN 55455, USA
| | - Jason R B Dyck
- Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, 87th Avenue and 112 Street, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
34
|
Wijnker PJ, Sequeira V, Kuster DW, van der Velden J. Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits. Antioxid Redox Signal 2019; 31:318-358. [PMID: 29490477 PMCID: PMC6602117 DOI: 10.1089/ars.2017.7236] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 02/23/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Significance: Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by left ventricular hypertrophy, diastolic dysfunction, and myocardial disarray. Disease onset occurs between 20 and 50 years of age, thus affecting patients in the prime of their life. HCM is caused by mutations in sarcomere proteins, the contractile building blocks of the heart. Despite increased knowledge of causal mutations, the exact path from genetic defect leading to cardiomyopathy is complex and involves additional disease hits. Recent Advances: Laboratory-based studies indicate that HCM development not only depends on the primary sarcomere impairment caused by the mutation but also on secondary disease-related alterations in the heart. Here we propose a vicious mutation-induced disease cycle, in which a mutation-induced energy depletion alters cellular metabolism with increased mitochondrial work, which triggers secondary disease modifiers that will worsen disease and ultimately lead to end-stage HCM. Critical Issues: Evidence shows excessive cellular reactive oxygen species (ROS) in HCM patients and HCM animal models. Oxidative stress markers are increased in the heart (oxidized proteins, DNA, and lipids) and serum of HCM patients. In addition, increased mitochondrial ROS production and changes in endogenous antioxidants are reported in HCM. Mutant sarcomeric protein may drive excessive levels of cardiac ROS via changes in cardiac efficiency and metabolism, mitochondrial activation and/or dysfunction, impaired protein quality control, and microvascular dysfunction. Future Directions: Interventions restoring metabolism, mitochondrial function, and improved ROS balance may be promising therapeutic approaches. We discuss the effects of current HCM pharmacological therapies and potential future therapies to prevent and reverse HCM. Antioxid. Redox Signal. 31, 318-358.
Collapse
Affiliation(s)
- Paul J.M. Wijnker
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Vasco Sequeira
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W.D. Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Advancements in highly active antiretroviral therapy (HAART) for human immunodeficiency virus (HIV) has led to increased survival, approaching that of the general population. These patients are at an increased risk for heart disease, specifically HIV-associated cardiomyopathy. RECENT FINDINGS Initially, HIV-associated cardiomyopathy was predominantly noted as systolic dysfunction, but diastolic dysfunction has become more common with increased use of HAART. The pathogenesis of HIV-associated cardiomyopathy involves direct viral infection, cytokine activity, focal myocarditis, HAART side effects, immune system dysregulation, and/or ischemia. If cardiomyopathy is diagnosed, an HIV patient should be started, or continued, on HAART and initiated on guideline-directed medical therapy (GDMT) per the ACC/AHA guidelines. The safety and efficacy of mechanical circulatory support and heart transplant in this population has been repeatedly demonstrated, but it remains less available compared with the general advanced heart failure population. SUMMARY The widespread use of HAART has led to prolonged survival in HIV patients to the point that cardiac disease has become the most common cause of death. Patients should be treated with HAART for HIV and GDMT for heart failure. Patients progressing to end-stage heart failure should be referred for advanced therapies to centers with experience treating HIV patients.
Collapse
|
36
|
Upregulation of UCP2 Expression Protects against LPS-Induced Oxidative Stress and Apoptosis in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2758262. [PMID: 31182990 PMCID: PMC6512061 DOI: 10.1155/2019/2758262] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/21/2019] [Accepted: 03/10/2019] [Indexed: 12/17/2022]
Abstract
Uncoupling protein 2 (UCP2) has a cardioprotective role under septic conditions, but the underlying mechanism remains unclear. This study aimed at investigating the effects of UCP2 on the oxidative stress and apoptosis of cardiomyocytes induced by lipopolysaccharide (LPS). First, LPS increased UCP2 expression in cardiomyocytes in a time-dependent manner. LPS increased the production of lactate dehydrogenase (LDH), reactive oxygen species (ROS), and malondialdehyde (MDA) and decreased the level of superoxide dismutase (SOD). However, UCP2 knockdown increased the LPS-induced cardiac injury and oxidative stress. In addition, LPS damaged the mitochondrial ultrastructure and led to the disruption of mitochondrial membrane potential (MMP), as well as the release of mitochondrial cytochrome c. UCP2 knockdown aggravated mitochondrial injury and the release of mitochondrial cytochrome c. LPS increased the protein levels of Bax and cleaved-caspase-3, decreased the protein level of Bcl-2, and upregulated the protein level of mitogen-activated protein kinase. However, upon UCP2 knockdown, the protein levels of Bax and cleaved-caspase-3 increased even further, and the protein level of Bcl-2 was further decreased. The protein level of phosphorylated p38 was also further enhanced. Thus, UCP2 protects against LPS-induced oxidative stress and apoptosis in cardiomyocytes.
Collapse
|
37
|
Stanisic J, Koricanac G, Kostic M, Stojiljkovic M, Culafic T, Romic S, Tepavcevic S. Low-intensity exercise in the prevention of cardiac insulin resistance-related inflammation and disturbances in NOS and MMP-9 regulation in fructose-fed ovariectomized rats. Appl Physiol Nutr Metab 2019; 44:1219-1229. [PMID: 30897341 DOI: 10.1139/apnm-2018-0785] [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] [Indexed: 12/25/2022]
Abstract
Exercise is important nonpharmacological treatment for improvement of insulin sensitivity in menopause. However, its effect on menopausal cardiac insulin resistance is needing further research. We investigated protective effects of low-intensity exercise on cardiac insulin signaling, inflammation, regulation of nitric oxide synthase (NOS) and matrix metalloproteinase 9 (MMP-9) in ovariectomized (OVX) Wistar rats, submitted to 10% fructose solution for 9 weeks. OVX rats were divided into control, sedentary fructose, and exercise fructose groups. Measurements of physical and biochemical characteristics were carried out to evaluate metabolic syndrome development. Messenger RNA and protein levels and phosphorylation of cardiac insulin signaling molecules, endothelial and inducible NOS (eNOS and iNOS), p65 subunit of nuclear factor κB (NFκB), tumor necrosis factor α (TNF-α), suppressor of cytokine signaling 3 (SOCS3), and MMP-9 were analyzed. Fructose increased insulin level, homeostasis model assessment (HOMA) index, and visceral adipose tissue weight, while low-intensity exercise prevented insulin level and HOMA index increase. Fructose also decreased cardiac pAkt (Ser473), peNOS (Ser1177) and increased insulin receptor substrate 1 (IRS1) phosphorylation at Ser307, pNFκB (Ser276) and NFκB and MMP-9 content, without any effect on iNOS, protein-tyrosine phosphatase 1B, TNF-α, and SOCS3. Exercise prevented changes in pIRS1 (Ser307), pAkt (Ser473), peNOS (Ser1177), pNFκB (Ser276), and NFκB expression. In addition, exercise increased pIRS1 (Tyr632), pAkt (Thr308), and eNOS expression. Low-intensity exercise prevented cardiac insulin signaling disarrangement in fructose-fed OVX rats and therefore eNOS dysfunction, as well as pro-inflammatory signaling activation, without effect on tissue remodeling, suggesting physical training as a way to reduce cardiovascular risk.
Collapse
Affiliation(s)
- Jelena Stanisic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Goran Koricanac
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Milan Kostic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Mojca Stojiljkovic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Tijana Culafic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Snjezana Romic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Snezana Tepavcevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| |
Collapse
|
38
|
Pharmacological strategies to lower crosstalk between nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. Biomed Pharmacother 2019; 111:1478-1498. [DOI: 10.1016/j.biopha.2018.11.128] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023] Open
|
39
|
De Nicola GF, Bassi R, Nichols C, Fernandez-Caggiano M, Golforoush PA, Thapa D, Anderson R, Martin ED, Verma S, Kleinjung J, Laing A, Hutchinson JP, Eaton P, Clark J, Marber MS. The TAB1-p38α complex aggravates myocardial injury and can be targeted by small molecules. JCI Insight 2018; 3:121144. [PMID: 30135318 PMCID: PMC6141180 DOI: 10.1172/jci.insight.121144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/05/2018] [Indexed: 11/26/2022] Open
Abstract
Inhibiting MAPK14 (p38α) diminishes cardiac damage in myocardial ischemia. During myocardial ischemia, p38α interacts with TAB1, a scaffold protein, which promotes p38α autoactivation; active p38α (pp38α) then transphosphorylates TAB1. Previously, we solved the X-ray structure of the p38α-TAB1 (residues 384–412) complex. Here, we further characterize the interaction by solving the structure of the pp38α-TAB1 (residues 1–438) complex in the active state. Based on this information, we created a global knock-in (KI) mouse with substitution of 4 residues on TAB1 that we show are required for docking onto p38α. Whereas ablating p38α or TAB1 resulted in early embryonal lethality, the TAB1-KI mice were viable and had no appreciable alteration in their lymphocyte repertoire or myocardial transcriptional profile; nonetheless, following in vivo regional myocardial ischemia, infarction volume was significantly reduced and the transphosphorylation of TAB1 was disabled. Unexpectedly, the activation of myocardial p38α during ischemia was only mildly attenuated in TAB1-KI hearts. We also identified a group of fragments able to disrupt the interaction between p38α and TAB1. We conclude that the interaction between the 2 proteins can be targeted with small molecules. The data reveal that it is possible to selectively inhibit signaling downstream of p38α to attenuate ischemic injury. Disrupting TAB1-p38α interaction in vivo has a protective effect during myocardial ischemia and can be achieved in vitro with small molecule inhibitors.
Collapse
Affiliation(s)
- Gian F De Nicola
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and.,The Randall Division, New Hunt's House, Guy's Campus, King's College London, United Kingdom
| | - Rekha Bassi
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - Charlie Nichols
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and.,The Randall Division, New Hunt's House, Guy's Campus, King's College London, United Kingdom
| | | | | | - Dibesh Thapa
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - Rhys Anderson
- The Randall Division, New Hunt's House, Guy's Campus, King's College London, United Kingdom
| | - Eva Denise Martin
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - Sharwari Verma
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - Jens Kleinjung
- Bioinformatics Facility, The Francis Crick Institute, London, United Kingdom
| | - Adam Laing
- Department of Immunobiology, King's College London, United Kingdom
| | - Jonathan P Hutchinson
- Platform Technologies and Science, GlaxoSmithKline, and.,Discovery Partnerships with Academia, GlaxoSmithKline, Medicines Research Centre, Stevenage, United Kingdom
| | - Philip Eaton
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - James Clark
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| | - Michael S Marber
- British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, and
| |
Collapse
|
40
|
Steiger D, Yokota T, Li J, Ren S, Minamisawa S, Wang Y. The serine/threonine-protein kinase/endoribonuclease IRE1α protects the heart against pressure overload-induced heart failure. J Biol Chem 2018; 293:9652-9661. [PMID: 29769316 DOI: 10.1074/jbc.ra118.003448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/04/2018] [Indexed: 12/26/2022] Open
Abstract
Heart failure is associated with induction of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). The serine/threonine protein kinase/endoribonuclease IRE1α is a key protein in ER stress signal transduction. IRE1α activity can induce both protective UPR and apoptotic downstream signaling events, but the specific role for IRE1α activity in the heart is unknown. A major aim of this study was to characterize the specific contribution of IRE1α in cardiac physiology and pathogenesis. We used both cultured myocytes and a transgenic mouse line with inducible and cardiomyocyte-specific IRE1α overexpression as experimental models to achieve targeted IRE1α activation. IRE1α expression induced a potent but transient ER stress response in cardiomyocytes and did not cause significant effects in the intact heart under normal physiological conditions. Furthermore, the IRE1α-activated transgenic heart responding to pressure overload exhibited preserved function and reduced fibrotic area, associated with increased adaptive UPR signaling and with blunted inflammatory and pathological gene expression. Therefore, we conclude that IRE1α induces transient ER stress signaling and confers a protective effect against pressure overload-induced pathological remodeling in the heart. To our knowledge, this report provides first direct evidence of a specific and protective role for IRE1α in the heart and reveals an interaction between ER stress signaling and inflammatory regulation in the pathologically stressed heart.
Collapse
Affiliation(s)
- DeAnna Steiger
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Tomohiro Yokota
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Jin Li
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Shuxun Ren
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| | - Susumu Minamisawa
- the Department of Cell Physiology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yibin Wang
- From the Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095 and
| |
Collapse
|
41
|
Morris G, Puri BK, Walder K, Berk M, Stubbs B, Maes M, Carvalho AF. The Endoplasmic Reticulum Stress Response in Neuroprogressive Diseases: Emerging Pathophysiological Role and Translational Implications. Mol Neurobiol 2018; 55:8765-8787. [PMID: 29594942 PMCID: PMC6208857 DOI: 10.1007/s12035-018-1028-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is the main cellular organelle involved in protein synthesis, assembly and secretion. Accumulating evidence shows that across several neurodegenerative and neuroprogressive diseases, ER stress ensues, which is accompanied by over-activation of the unfolded protein response (UPR). Although the UPR could initially serve adaptive purposes in conditions associated with higher cellular demands and after exposure to a range of pathophysiological insults, over time the UPR may become detrimental, thus contributing to neuroprogression. Herein, we propose that immune-inflammatory, neuro-oxidative, neuro-nitrosative, as well as mitochondrial pathways may reciprocally interact with aberrations in UPR pathways. Furthermore, ER stress may contribute to a deregulation in calcium homoeostasis. The common denominator of these pathways is a decrease in neuronal resilience, synaptic dysfunction and even cell death. This review also discusses how mechanisms related to ER stress could be explored as a source for novel therapeutic targets for neurodegenerative and neuroprogressive diseases. The design of randomised controlled trials testing compounds that target aberrant UPR-related pathways within the emerging framework of precision psychiatry is warranted.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| | - Ken Walder
- The Centre for Molecular and Medical Research, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
- Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Florey Institute for Neuroscience and Mental Health, Melbourne, Australia
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Chelmsford, UK
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
42
|
Guo CA, Guo S. Insulin receptor substrate signaling controls cardiac energy metabolism and heart failure. J Endocrinol 2017; 233:R131-R143. [PMID: 28381504 PMCID: PMC9675292 DOI: 10.1530/joe-16-0679] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
The heart is an insulin-dependent and energy-consuming organ in which insulin and nutritional signaling integrates to the regulation of cardiac metabolism, growth and survival. Heart failure is highly associated with insulin resistance, and heart failure patients suffer from the cardiac energy deficiency and structural and functional dysfunction. Chronic pathological conditions, such as obesity and type 2 diabetes mellitus, involve various mechanisms in promoting heart failure by remodeling metabolic pathways, modulating cardiac energetics and impairing cardiac contractility. Recent studies demonstrated that insulin receptor substrates 1 and 2 (IRS-1,-2) are major mediators of both insulin and insulin-like growth factor-1 (IGF-1) signaling responsible for myocardial energetics, structure, function and organismal survival. Importantly, the insulin receptor substrates (IRS) play an important role in the activation of the phosphatidylinositide-3-dependent kinase (PI-3K) that controls Akt and Foxo1 signaling cascade, regulating the mitochondrial function, cardiac energy metabolism and the renin-angiotensin system. Dysregulation of this branch in signaling cascades by insulin resistance in the heart through the endocrine system promotes heart failure, providing a novel mechanism for diabetic cardiomyopathy. Therefore, targeting this branch of IRS→PI-3K→Foxo1 signaling cascade and associated pathways may provide a fundamental strategy for the therapeutic and nutritional development in control of metabolic and cardiovascular diseases. In this review, we focus on insulin signaling and resistance in the heart and the role energetics play in cardiac metabolism, structure and function.
Collapse
Affiliation(s)
- Cathy A Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Shaodong Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
43
|
Paricalcitol Attenuates Cardiac Fibrosis and Expression of Endothelial Cell Transition Markers in Isoproterenol-Induced Cardiomyopathic Rats. Crit Care Med 2017; 44:e866-74. [PMID: 27065465 DOI: 10.1097/ccm.0000000000001736] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Acute cardiomyopathy is a health problem worldwide. Few studies have shown an association between acute cardiomyopathy and low vitamin D status. Paricalcitol, a vitamin D receptor activator, clinically benefits patients with advanced kidney disease. The effect of paricalcitol supplement on cardiac remodeling in cardiomyopathic rats is unknown. This experimental study investigated the effect of paricalcitol in rats with cardiomyopathy induced by isoproterenol. DESIGN Prospective, randomized, controlled experimental study. SETTING Hospital-affiliated animal research institution. SUBJECTS Eight-week-old male Wistar-Kyoto rats. INTERVENTIONS Male Wistar-Kyoto rats were first injected intraperitoneally with isoproterenol to create a rat model of acute cardiomyopathy. Then paricalcitol was administered intraperitoneally to isoproterenol-injected rats at a dosage of 200 ng three times a week for 3 weeks. Relevant cardiomyopathy-related variables were measured regularly in three groups of rats, controls, isoproterenol, and isoproterenol plus paricalcitol. Rat hearts were obtained for evaluation of cardiac fibrosis using Masson trichrome staining and commercially available software, and evaluation of cell transition using immunofluorescence staining analysis. MEASUREMENTS AND MAIN RESULTS Isoproterenol infusions generated significant cardiac fibrosis (p < 0.001). Subsequent paricalcitol treatment attenuated the isoproterenol-induced cardiac fibrosis (p = 0.006). Fluorescence showed colocalization of endothelial and fibroblast cell markers (cluster differentiation 31 and α-smooth muscle actin, respectively) in the isoproterenol-treated hearts. Paricalcitol injections attenuated the isoproterenol-induced fluorescence intensity of two cell markers (p < 0.01). CONCLUSIONS Paricalcitol injections may ameliorate isoproterenol-induced cardiac fibrosis possibly through regulating cell transition.
Collapse
|
44
|
Yue-Chun L, Guang-Yi C, Li-Sha G, Chao X, Xinqiao T, Cong L, Xiao-Ya D, Xiangjun Y. The Protective Effects of Ivabradine in Preventing Progression from Viral Myocarditis to Dilated Cardiomyopathy. Front Pharmacol 2016; 7:408. [PMID: 27847478 PMCID: PMC5088506 DOI: 10.3389/fphar.2016.00408] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 10/14/2016] [Indexed: 12/04/2022] Open
Abstract
To study the beneficial effects of ivabradine in dilated cardiomyopathy (DCM) mice, which evolved from coxsackievirus B3-induced chronic viral myocarditis. Four-to-five-week-old male balb/c mice were inoculated intraperitoneally with coxsackievirus B3 (Strain Nancy) on days 1, 14, and 28. The day of the first virus inoculation was defined as day 1. Thirty-five days later, the surviving chronic viral myocarditis mice were divided randomly into two groups, a treatment group and an untreated group. Ivabradine was administered by gavage for 30 consecutive days in the treatment group, and the untreated group was administered normal saline. Masson’s trichrome stain was used to evaluate the fibrosis degree in myocardial tissue. The expression levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), collagen I, collagen III and p38-MAPK signaling pathway proteins were detected by Western blot. Electrocardiogram was used to investigate the heart rate and rhythm. The thickness of the ventricular septum and left ventricular posterior wall, left ventricular end diastolic dimension, left ventricular end systolic dimension, left ventricular ejection fractions and fractional shortening were studied by echocardiography. Compared with the untreated chronic viral myocarditis mice, ivabradine significantly increased the survival rate, attenuated the myocardial lesions and fibrosis, improved the impairment of the left ventricular function, diminished the heart dimension, decreased the production of collagen I and collagen III, reduced the expression of the proinflammatory cytokines TNF-α, IL-1β, and IL-6, and lowered the production of phospho-p38 MAPK. The findings indicate the therapeutic effect of ivabradine in preventing the progression from viral myocarditis to DCM in mice with chronic viral myocarditis induced by coxsackievirus B3, is associated with inhibition of the p38 MAPK pathway, downregulated inflammatory responses and decreased collagen expression. Ivabradine appears a promising approach for the treatment of patients with viral myocarditis.
Collapse
Affiliation(s)
- Li Yue-Chun
- Department of Cardiology, First Affiliated Hospital of Soochow UniversitySuzhou, China; Department of Cardiology, Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Chen Guang-Yi
- Department of Cardiology, Second Affiliated Hospital of Wenzhou Medical University Wenzhou, China
| | - Ge Li-Sha
- Department of Pediatrics, Second Affiliated Hospital of Wenzhou Medical University Wenzhou, China
| | - Xing Chao
- Department of Clinical Laboratory, Second Affiliated Hospital of Wenzhou Medical University Wenzhou, China
| | - Tian Xinqiao
- Department of Ultrasonography, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou China
| | - Lin Cong
- Department of Cardiology, Second Affiliated Hospital of Wenzhou Medical University Wenzhou, China
| | - Dai Xiao-Ya
- Department of Cardiology, Second Affiliated Hospital of Wenzhou Medical University Wenzhou, China
| | - Yang Xiangjun
- Department of Cardiology, First Affiliated Hospital of Soochow University Suzhou, China
| |
Collapse
|
45
|
Luo M, Luo P, Zhang Z, Payne K, Watson S, Wu H, Tan Y, Ding Y, Sun W, Yin X, Zhang X, Liu G, Wintergerst K, Miao L, Cai L. Zinc delays the progression of obesity-related glomerulopathy in mice via down-regulating P38 MAPK-mediated inflammation. Obesity (Silver Spring) 2016; 24:1244-1256. [PMID: 27028368 PMCID: PMC7218660 DOI: 10.1002/oby.21463] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/14/2015] [Accepted: 01/02/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Obesity, particularly child obesity, is one of the most common public health problems in the world and raises the risk of end-stage renal disease. Zinc (Zn) is essential for multiple organs in terms of normal structure and function; however, effects of Zn deficiency or supplementation among young individuals with obesity have not been well studied. METHODS Weaned mice were fed high-fat diets (HFD) with varied contents of Zn (Zn deficient, adequate, and supplemented) for 3 or 6 months. This study examined associations between renal pathogenesis and dietary Zn levels, specifically assessing inflammatory pathways by utilizing P38 MAPK inhibitor SB203580. RESULTS HFD feeding induced typical syndromes of obesity-related renal disorders, which worsened by Zn marginal deficiency. The progression of obesity-related renal disorders was delayed by Zn supplementation. HFD induced renal inflammation, reflected by increased P38 MAPK phosphorylation along with increases of inflammatory cytokines MCP-1, IL-1β, IL-6, and TNF-α. P38 MAPK inhibition prevented renal pathological changes in mice fed with HFD and HFD/Zn deficiency. CONCLUSIONS P38 MAPK mediated the renal inflammatory responses, which played a central role in the pathogenesis of HFD-induced renal disorders. Zn could delay the progression of obesity-related kidney disease by down-regulating P38 MAPK-mediated inflammation.
Collapse
Affiliation(s)
- Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
- Department of Pediatrics, The University of Louisville, Kentucky, USA
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiguo Zhang
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kristen Payne
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- Department of Internal Medicine, The Marshall University Joan C. Edwards School of Medicine, Huntington, West Virginia, USA
| | - Sara Watson
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- Wendy L. Novak Diabetes Care Center, The University of Louisville School of Medicine, Kentucky, USA
| | - Hao Wu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
- Department of Pediatrics, The University of Louisville, Kentucky, USA
| | - Yi Tan
- Department of Pediatrics, The University of Louisville, Kentucky, USA
| | - Yushuang Ding
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinmin Yin
- Department of Chemistry, The University of Louisville, Kentucky, USA
| | - Xiang Zhang
- Department of Chemistry, The University of Louisville, Kentucky, USA
| | - Gilbert Liu
- Department of Pediatrics, The University of Louisville, Kentucky, USA
| | - Kupper Wintergerst
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- Wendy L. Novak Diabetes Care Center, The University of Louisville School of Medicine, Kentucky, USA
| | - Lining Miao
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lu Cai
- Department of Pediatrics, The University of Louisville, Kentucky, USA
- Wendy L. Novak Diabetes Care Center, The University of Louisville School of Medicine, Kentucky, USA
| |
Collapse
|
46
|
TGF-β receptor type II costameric localization in cardiomyocytes and host cell TGF-β response is disrupted by Trypanosoma cruzi infection. Parasitology 2016; 143:704-15. [DOI: 10.1017/s0031182016000299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYTransforming growth factor beta (TGF-β) cytokine is involved in Chagas disease establishment and progression. Since Trypanosoma cruzi can modulate host cell receptors, we analysed the TGF-β receptor type II (TβRII) expression and distribution during T. cruzi – cardiomyocyte interaction. TβRII immunofluorescent staining revealed a striated organization in cardiomyocytes, which was co-localized with vinculin costameres and enhanced (38%) after TGF-β treatment. Cytochalasin D induced a decrease of 45·3% in the ratio of cardiomyocytes presenting TβRII striations, demonstrating an association of TβRII with the cytoskeleton. Western blot analysis showed that cytochalasin D significantly inhibited Smad 2 phosphorylation and fibronectin stimulation after TGF-β treatment in cardiomyocytes. Trypanosoma cruzi infection elicited a decrease of 79·8% in the frequency of cardiomyocytes presenting TβRII striations, but did not interfere significantly in its expression. In addition, T. cruzi-infected cardiomyocytes present a lower response to exogenous TGF-β, showing no enhancement of TβRII striations and a reduction of phosphorylated Smad 2, with no significant difference in TβRII expression when compared to uninfected cells. Together, these results suggest that the co-localization of TβRII with costameres is important in activating the TGF-β signalling cascade, and that T. cruzi-derived cytoskeleton disorganization could result in altered or low TGF-β response in infected cardiomyocytes.
Collapse
|
47
|
Spindler MP, Ho AM, Tridgell D, McCulloch-Olson M, Gersuk V, Ni C, Greenbaum C, Sanda S. Acute hyperglycemia impairs IL-6 expression in humans. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:91-7. [PMID: 27042306 PMCID: PMC4768063 DOI: 10.1002/iid3.97] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022]
Abstract
Normal glucose metabolism is critical to immune function but the effects of short‐term hyperglycemia on immunity are not well described. To study this phenomenon, we induced hyperglycemia in healthy subjects for 2 h with intravenous dextrose and octreotide. An RNA‐seq analysis of whole blood RNA demonstrated alterations in multiple immune pathways and transcripts during acute hyperglycemia including decreased transcription of IL‐6, an important component of both innate and adaptive immune responses. Additional in vitro studies of human peripheral blood mononuclear cells (PBMCs) exposed to high glucose confirmed decreased IL‐6 expression, most prominently in CD14+CD16+ intermediate monocytes. Hyperglycemia also reduced IL‐17A expression suggesting further impairment of immune responses during acute hyperglycemia. These findings demonstrate multiple defective immune responses in acute hyperglycemia and suggest a novel role for intermediate monocytes as metabolically sensitive innate immune cells.
Collapse
Affiliation(s)
- Matthew P Spindler
- The Diabetes Center UCSF School of Medicine San Francicsco California 94143 USA
| | - Alvin M Ho
- The Diabetes Center UCSF School of Medicine San Francicsco California 94143 USA
| | - David Tridgell
- The Diabetes Research Program Benaroya Research Institute Seattle Washington 98040 USA
| | - Marli McCulloch-Olson
- The Diabetes Research Program Benaroya Research Institute Seattle Washington 98040 USA
| | - Vivian Gersuk
- The Diabetes Research Program Benaroya Research Institute Seattle Washington 98040 USA
| | - Chester Ni
- The Diabetes Research Program Benaroya Research Institute Seattle Washington 98040 USA
| | - Carla Greenbaum
- The Diabetes Research Program Benaroya Research Institute Seattle Washington 98040 USA
| | - Srinath Sanda
- The Diabetes Center UCSF School of Medicine San Francicsco California 94143 USA
| |
Collapse
|
48
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
49
|
Evaluation of Proinflammatory Prognostic Biomarkers for Fabry Cardiomyopathy With Enzyme Replacement Therapy. Can J Cardiol 2015; 32:1221.e1-1221.e9. [PMID: 26919792 DOI: 10.1016/j.cjca.2015.10.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Fabry disease (FD) causes progressive glycosphingolipid accumulation and damage in various organs, and several proinflammatory processes may be involved in this disease. Enzyme replacement therapy (ERT) can reduce the severity of Fabry cardiomyopathy (FC), but whether ERT could attenuate proinflammatory cytokines in FC remains unclear. In this study, we attempted to evaluate the efficacy of ERT on proinflammatory cytokines and vascular cell adhesion biomarkers. METHODS We enrolled 25 patients with FC and administered ERT to them according to the present clinical guideline. We analyzed and compared echocardiographic and blood examination results between 25 patients with FD without left ventricular hypertrophy (LVH), 25 patients with FC with LVH who were receiving ERT, and 25 healthy age- and sex-matched controls. The parameters of cardiac function at baseline and 12 months after ERT were assessed through echocardiography, and the expression profiles of proinflammatory biomarkers were determined. RESULTS Left ventricular mass (LVM), LVM index (LVMI), interventricular septal thickness at diastole, and serum levels of globotriaosylsphingosine (Gb3) were elevated in patients with FC. Meanwhile, several proinflammatory cytokines, including interleukin (IL)-6, IL-2, IL-1b, tumor necrosis factor-α, intercellular adhesion molecule, soluble vascular cell adhesion molecule, and monocyte chemoattractant protein-1 (MCP-1) were concomitantly increased. ERT significantly reduced these transthoracic echocardiographic parameters and lyso-Gb3 and proinflammatory cytokine levels. The changes in IL-6, MCP-1, and lyso-Gb3 levels were positively correlated with the change in LVMI. CONCLUSIONS Our study has revealed that proinflammatory biomarkers, particularly IL-6 and MCP-1, may represent effective biomarkers for evaluating ERT outcomes in patients with FC.
Collapse
|
50
|
Lucas A, Mialet-Perez J, Daviaud D, Parini A, Marber MS, Sicard P. Gadd45γ regulates cardiomyocyte death and post-myocardial infarction left ventricular remodelling. Cardiovasc Res 2015; 108:254-67. [PMID: 26370247 DOI: 10.1093/cvr/cvv219] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/08/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS Post-infarction remodelling is accompanied and influenced by perturbations in mitogen-activated protein kinase (MAPK) signalling. The growth arrest and DNA-damage-inducible 45 (Gadd45) proteins are small acidic proteins involved in DNA repair and modulation of MAPK activity. Little is known about the role of Gadd45 in the heart. Here, we explored the potential contribution of Gadd45 gamma (γ) isoform to the acute and late phase of heart failure (HF) after myocardial infarction (MI) and determined the mechanisms underlying Gadd45γ actions. METHODS AND RESULTS The Gadd45γ isoform is up-regulated in murine cardiomyocytes subjected to simulated ischaemia and in the mouse heart during MI. To mimic the situation observed during MI, we enhanced Gadd45γ content in cardiomyocytes with a single injection of an adeno-associated viral (AAV9) vector encoding Gadd45γ under the cTNT promoter. Gadd45γ overexpression induces cardiomyocyte apoptosis, fibrosis, left ventricular dysfunction, and HF. On the other hand, genetic deletion of Gadd45γ in knockout mice confers resistance to ischaemic injury, at least in part by limiting cardiomyocyte apoptosis. Mechanistically, Gadd45γ activates receptor-interacting protein 1 (RIP1) and caspase-8 in a p38 MAPK-dependent manner to promote cardiomyocyte death. CONCLUSION This work is the first to demonstrate that Gadd45γ accumulation during MI promotes the development and persistence of HF by inducing cardiomyocyte apoptosis in a p38 MAPK-dependent manner. We clearly identify Gadd45γ as a therapeutic target in the development of HF.
Collapse
Affiliation(s)
- Alexandre Lucas
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, 1 Avenue Jean Poulhes, 31432 Toulouse, France University Paul Sabatier, CHU of Toulouse, 31432 Toulouse, France
| | - Jeanne Mialet-Perez
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, 1 Avenue Jean Poulhes, 31432 Toulouse, France University Paul Sabatier, CHU of Toulouse, 31432 Toulouse, France
| | - Danièle Daviaud
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, 1 Avenue Jean Poulhes, 31432 Toulouse, France University Paul Sabatier, CHU of Toulouse, 31432 Toulouse, France
| | - Angelo Parini
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, 1 Avenue Jean Poulhes, 31432 Toulouse, France University Paul Sabatier, CHU of Toulouse, 31432 Toulouse, France
| | - Michael S Marber
- Cardiovascular Division, King's College London, The Rayne Institute, St. Thomas' Hospital, London, UK
| | - Pierre Sicard
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, 1 Avenue Jean Poulhes, 31432 Toulouse, France University Paul Sabatier, CHU of Toulouse, 31432 Toulouse, France
| |
Collapse
|