1
|
Du C, Wang S, Shi X, Jing P, Wang H, Wang L. Identification of senescence related hub genes and potential therapeutic compounds for dilated cardiomyopathy via comprehensive transcriptome analysis. Comput Biol Med 2024; 179:108901. [PMID: 39029429 DOI: 10.1016/j.compbiomed.2024.108901] [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: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a common cause of heart failure. However, the role of cellular senescence in DCM has not been fully elucidated. Here, we aimed to investigate senescence in DCM, identify senescence related characteristic genes, and explore the potential small molecule compounds for DCM treatment. METHODS DCM-associated datasets and senescence-related genes were respectively obtained from Gene Expression Omnibus (GEO) database and CellAge database. The characteristic genes were identified through methods including weighted gene co-expression network analysis (WGCNA), least absolute shrinkage and selection operator (LASSO), and random forest. The expression of characteristic genes was verified in the mouse DCM model. Moreover, the CIBERSORT algorithm was applied to analyze immune characteristics of DCM. Finally, several therapeutic compounds were predicted by CMap analysis, and the potential mechanism of chlorogenic acid (CGA) was investigated by molecular docking and molecular dynamics simulation. RESULTS Three DCM- and senescence-related characteristic genes (MME, GNMT and PLA2G2A) were ultimately identified through comprehensive transcriptome analysis, and were experimentally verified in the doxorubicin induced mouse DCM. Meanwhile, the established diagnostic model, derived from dataset analysis, showed ideal diagnostic performance for DCM. Immune cell infiltration analysis suggested dysregulation of inflammation in DCM, and the characteristic genes were significantly associated with invasive immune cells. Finally, based on the specific gene expression profile of DCM, several potential therapeutic compounds were predicted through CMap analysis. In addition, molecular docking and molecular dynamics simulations suggested that CGA could bind to the active pocket of MME protein. CONCLUSION Our study presents three characteristic genes (MME, PLA2G2A, and GNMT) and a novel senescence-based diagnostic nomogram, and discusses potential therapeutic compounds, providing new insights into the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Chong Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Sibo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinying Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Peng Jing
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Liansheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
2
|
Dos Santos Jorge Sousa K, de Souza A, de Almeida Cruz M, de Lima LE, do Espirito Santo G, Amaral GO, Granito RN, Renno AC. 3D printed scaffolds of biosilica and spongin from marine sponges: analysis of genotoxicity and cytotoxicity for bone tissue repair. Bioprocess Biosyst Eng 2024; 47:1483-1498. [PMID: 38869621 DOI: 10.1007/s00449-024-03042-z] [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: 02/08/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Biosilica (BS) and spongin (SPG) from marine sponges are highlighted for their potential to promote bone regeneration. Moreover, 3D printing is introduced as a technology for producing bone grafts with optimized porous structures, allowing for better cell attachment, proliferation, and differentiation. Thus, this study aimed to characterize the BS and BS/SPG 3D printed scaffolds and to evaluate the biological effects in vitro. The scaffolds were printed using an ink containing 4 wt.% of sodium alginate. The physicochemical characteristics of BS and BS/SPG 3D printed scaffolds were analyzed by SEM, EDS, FTIR, porosity, evaluation of mass loss, and pH measurement. For in vitro analysis, the cellular viability of the MC3T3-E1 cell lineage was assessed using the AlamarBlue® assay and confocal microscopy, while genotoxicity and mineralization potential were evaluated through the micronucleus assay and Alizarin Red S, respectively. SEM analysis revealed spicules in BS, the fibrillar structure of SPG, and material degradation over the immersion period. FTIR indicated peaks corresponding to silicon oxide in BS samples and carbon oxide and amine in SPG samples. BS-SPG scaffolds exhibited higher porosity, while BS scaffolds displayed greater mass loss. pH measurements indicated a significant decrease induced by BS, which was mitigated by SPG over the experimental periods. In vitro studies demonstrated the biocompatibility and non-cytotoxicity of scaffold extracts. .Also, the scaffolds promoted cellular differentiation. The micronucleus test further confirmed the absence of genotoxicity. These findings suggest that 3D printed BS and BS/SPG scaffolds may possess desirable morphological and physicochemical properties, indicating in vitro biocompatibility.
Collapse
Affiliation(s)
- Karolyne Dos Santos Jorge Sousa
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil.
| | - Amanda de Souza
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Matheus de Almeida Cruz
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Lindiane Eloisa de Lima
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Giovanna do Espirito Santo
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Gustavo Oliva Amaral
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Renata Neves Granito
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| | - Ana Claudia Renno
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP, 11015020, Brazil
| |
Collapse
|
3
|
Elias-Llumbet A, Sharmin R, Berg-Sorensen K, Schirhagl R, Mzyk A. The Interplay between Mechanoregulation and ROS in Heart Physiology, Disease, and Regeneration. Adv Healthc Mater 2024; 13:e2400952. [PMID: 38962858 DOI: 10.1002/adhm.202400952] [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: 03/13/2024] [Revised: 05/16/2024] [Indexed: 07/05/2024]
Abstract
Cardiovascular diseases are currently the most common cause of death in developed countries. Due to lifestyle and environmental factors, this problem is only expected to increase in the future. Reactive oxygen species (ROS) are a key player in the onset of cardiovascular diseases but also have important functions in healthy cardiac tissue. Here, the interplay between ROS generation and cardiac mechanical forces is shown, and the state of the art and a perspective on future directions are discussed. To this end, an overview of what is currently known regarding ROS and mechanosignaling at a subcellular level is first given. There the role of ROS in mechanosignaling as well as the interplay between both factors in specific organelles is emphasized. The consequences at a larger scale across the population of heart cells are then discussed. Subsequently, the roles of ROS in embryogenesis, pathogenesis, and aging are further discussed, exemplifying some aspects of mechanoregulation. Finally, different models that are currently in use are discussed to study the topics above.
Collapse
Affiliation(s)
- Arturo Elias-Llumbet
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
- Laboratory of Genomic of Germ Cells, Biomedical Sciences Institute, Faculty of Medicine, University of Chile, Independencia, Santiago, 1027, Chile
| | - Rokshana Sharmin
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | | | - Romana Schirhagl
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | - Aldona Mzyk
- DTU Health Tech, Ørsteds Plads Bldg 345C, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
4
|
He K, Zhang H, Tan B, Song C, Liang Z, Zhang L, Tian D, Xiao L, Xue H, Guo Q, Teng X, Jin S, An C, Wu Y. Hydrogen Sulfide Ameliorates Heart Aging by Downregulating Matrix Metalloproteinase-9. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07586-w. [PMID: 38884920 DOI: 10.1007/s10557-024-07586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Aging contributes significantly to cardiovascular diseases and cardiac dysfunction, leading to the upregulation of matrix metalloproteinase-9 (MMP-9) in the heart and a significant decrease in hydrogen sulfide (H2S) content, coupled with impaired cardiac diastolic function. This study explores whether supplementing exogenous hydrogen sulfide during aging ameliorates the decline in H2S concentration in the heart, suppresses MMP-9 expression, and improves the age-associated impairment in cardiac morphology and function. METHODS We collected plasma from healthy individuals of different ages to determine the relationship between aging and H2S and MMP-9 levels through Elisa detection and liquid chromatography-tandem mass spectrometry (LC/MC) detection of plasma H2S content. Three-month-old mice were selected as the young group, while 18-month-old mice were selected as the old group, and sodium hydrosulfide (NaHS) was injected intraperitoneally from 15 months old until 18 months old as the old + NaHS group. Plasma MMP-9 content was detected using Elisa, plasma H2S content, cardiac H2S content, and cystathionine gamma-lyase (CSE) activity were detected using LC/MC, and cardiac function was detected using echocardiography. Heart structure was assessed using hematoxylin and eosin staining, Masone staining was used to detect the degree of cardiac fibrosis, while western blot was used to detect the expression of MMP-9, CSE, and aging marker proteins. Knockdown of MMP-9 and CSE in H9c2 cells using small interfering RNA was carried out to determine the upstream-downstream relationship between MMP-9 and CSE. RESULTS H2S content in the plasma of healthy individuals decreases with escalating age, whereas MMP-9 level rises with age progression. Aging leads to a decrease in H2S levels in the heart and plasma of mice, severe impairment of cardiac diastolic function, interstitial relaxation, and fibrosis of the heart. Supplementing with exogenous H2S can improve these phenomena. CONCLUSION H2S maintains the structure and function of the heart by inhibiting the expression of MMP-9 during the aging process.
Collapse
Affiliation(s)
- Kaichuan He
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, 050017, Hebei, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Chengqing Song
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Zihui Liang
- Clinical Practice Teaching Department, Hebei Medical University, 050017, Hebei, China
| | - Lixia Zhang
- Department of Medical Laboratory, Hebei Children's Hospital, 050017, Hebei, China
| | - Danyang Tian
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Lin Xiao
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Hongmei Xue
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Cuixia An
- Department of Psychiatry, the First Hospital of Hebei Medical University, 050031, Hebei, China.
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, 050017, Hebei, China.
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, 050017, Hebei, China.
- Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017, Hebei, China.
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017, Hebei, China.
| |
Collapse
|
5
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
6
|
Park S, Laskow TC, Chen J, Guha P, Dawn B, Kim D. Microphysiological systems for human aging research. Aging Cell 2024; 23:e14070. [PMID: 38180277 PMCID: PMC10928588 DOI: 10.1111/acel.14070] [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: 06/30/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in microphysiological systems (MPS), also known as organs-on-a-chip (OoC), enable the recapitulation of more complex organ and tissue functions on a smaller scale in vitro. MPS therefore provide the potential to better understand human diseases and physiology. To date, numerous MPS platforms have been developed for various tissues and organs, including the heart, liver, kidney, blood vessels, muscle, and adipose tissue. However, only a few studies have explored using MPS platforms to unravel the effects of aging on human physiology and the pathogenesis of age-related diseases. Age is one of the risk factors for many diseases, and enormous interest has been devoted to aging research. As such, a human MPS aging model could provide a more predictive tool to understand the molecular and cellular mechanisms underlying human aging and age-related diseases. These models can also be used to evaluate preclinical drugs for age-related diseases and translate them into clinical settings. Here, we provide a review on the application of MPS in aging research. First, we offer an overview of the molecular, cellular, and physiological changes with age in several tissues or organs. Next, we discuss previous aging models and the current state of MPS for studying human aging and age-related conditions. Lastly, we address the limitations of current MPS and present future directions on the potential of MPS platforms for human aging research.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical EngineeringUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Thomas C. Laskow
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jingchun Chen
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Prasun Guha
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
- School of Life SciencesUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Deok‐Ho Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
7
|
Luan Y, Zhu X, Jiao Y, Liu H, Huang Z, Pei J, Xu Y, Yang Y, Ren K. Cardiac cell senescence: molecular mechanisms, key proteins and therapeutic targets. Cell Death Discov 2024; 10:78. [PMID: 38355681 PMCID: PMC10866973 DOI: 10.1038/s41420-023-01792-5] [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: 09/04/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Cardiac aging, particularly cardiac cell senescence, is a natural process that occurs as we age. Heart function gradually declines in old age, leading to continuous heart failure, even in people without a prior history of heart disease. To address this issue and improve cardiac cell function, it is crucial to investigate the molecular mechanisms underlying cardiac senescence. This review summarizes the main mechanisms and key proteins involved in cardiac cell senescence. This review further discusses the molecular modulators of cellular senescence in aging hearts. Furthermore, the discussion will encompass comprehensive descriptions of the key drugs, modes of action and potential targets for intervention in cardiac senescence. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of cardiac senescence, this review seeks to provide a fresh perspective and important theoretical foundations for the development of drugs targeting this condition.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Xiaofan Zhu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People's Hospital, Zhengzhou, 450052, P. R. China
| | - Yawei Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
8
|
Kwon HY, Yoon Y, Hong JE, Rhee KJ, Sohn JH, Jung PY, Kim MY, Baik SK, Ryu H, Eom YW. Role of TGF-β and p38 MAPK in TSG-6 Expression in Adipose Tissue-Derived Stem Cells In Vitro and In Vivo. Int J Mol Sci 2023; 25:477. [PMID: 38203646 PMCID: PMC10778696 DOI: 10.3390/ijms25010477] [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: 11/13/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Mesenchymal stem cells (MSCs) regulate immune cell activity by expressing tumor necrosis factor-α (TNF-α)-stimulated gene 6 (TSG-6) in inflammatory environments; however, whether anti-inflammatory responses affect TSG-6 expression in MSCs is not well understood. Therefore, we investigated whether transforming growth factor-β (TGF-β) regulates TSG-6 expression in adipose tissue-derived stem cells (ASCs) and whether effective immunosuppression can be achieved using ASCs and TGF-β signaling inhibitor A83-01. TGF-β significantly decreased TSG-6 expression in ASCs, but A83-01 and the p38 inhibitor SB202190 significantly increased it. However, in septic C57BL/6 mice, A83-01 further reduced the survival rate of the lipopolysaccharide (LPS)-treated group and ASC transplantation did not improve the severity induced by LPS. ASC transplantation alleviated the severity of sepsis induced by LPS+A83-01. In co-culture of macrophages and ASCs, A83-01 decreased TSG-6 expression whereas A83-01 and SB202190 reduced Cox-2 and IDO-2 expression in ASCs. These results suggest that TSG-6 expression in ASCs can be regulated by high concentrations of pro-inflammatory cytokines in vitro and in vivo, and that A83-01 and SB202190 can reduce the expression of immunomodulators in ASCs. Therefore, our data suggest that co-treatment of ASCs with TGF-β or p38 inhibitors is not adequate to modulate inflammation.
Collapse
Affiliation(s)
- Hye Youn Kwon
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Yongdae Yoon
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (J.-E.H.); (K.-J.R.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (J.-E.H.); (K.-J.R.)
| | - Joon Hyung Sohn
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| | - Pil Young Jung
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Moon Young Kim
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Soon Koo Baik
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Hoon Ryu
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Young Woo Eom
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
| |
Collapse
|
9
|
Zhang Y, He Y, Liu S, Deng L, Zuo Y, Huang K, Liao B, Li G, Feng J. SGLT2 Inhibitors in Aging-Related Cardiovascular Disease: A Review of Potential Mechanisms. Am J Cardiovasc Drugs 2023; 23:641-662. [PMID: 37620652 DOI: 10.1007/s40256-023-00602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/26/2023]
Abstract
Population aging combined with higher susceptibility to cardiovascular diseases in older adults is increasing the incidence of conditions such as atherosclerosis, myocardial infarction, heart failure, myocardial hypertrophy, myocardial fibrosis, arrhythmia, and hypertension. sodium-glucose cotransporter 2 inhibitors (SGLT2i) were originally developed as a novel oral drug for patients with type 2 diabetes mellitus. Unexpectedly, recent studies have shown that, beyond their effect on hyperglycemia, SGLT2i also have a variety of beneficial effects on cardiovascular disease. Experimental models of cardiovascular disease have shown that SGLT2i ameliorate the process of aging-related cardiovascular disease by inhibiting inflammation, reducing oxidative stress, and reversing endothelial dysfunction. In this review, we discuss the role of SGLT2i in aging-related cardiovascular disease and propose the use of SGLT2i to prevent and treat these conditions in older adults.
Collapse
Affiliation(s)
- Yali Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Siqi Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yumei Zuo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Department of Cardiac Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guang Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
10
|
Tocantins C, Martins JD, Rodrigues ÓM, Grilo LF, Diniz MS, Stevanovic-Silva J, Beleza J, Coxito P, Rizo-Roca D, Santos-Alves E, Rios M, Carvalho L, Moreno AJ, Ascensão A, Magalhães J, Oliveira PJ, Pereira SP. Metabolic mitochondrial alterations prevail in the female rat heart 8 weeks after exercise cessation. Eur J Clin Invest 2023; 53:e14069. [PMID: 37525474 DOI: 10.1111/eci.14069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/15/2023] [Accepted: 06/30/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND The consumption of high-caloric diets strongly contributes to the development of non-communicable diseases (NCDs), including cardiovascular disease, the leading cause of mortality worldwide. Exercise (along with diet intervention) is one of the primary non-pharmacological approaches to promote a healthier lifestyle and counteract the rampant prevalence of NCDs. The present study evaluated the effects of exercise cessation after a short period training on the cardiac metabolic and mitochondrial function of female rats. METHODS Seven-week-old female Sprague-Dawley rats were fed a control or a high-fat, high-sugar (HFHS) diet and, after 7 weeks, the animals were kept on a sedentary lifestyle or submitted to endurance exercise for 3 weeks (6 days per week, 20-60 min/day). The cardiac samples were analysed 8 weeks after exercise cessation. RESULTS The consumption of the HFHS diet triggered impaired glucose tolerance, whereas the HFHS diet and physical exercise resulted in different responses in plasma adiponectin and leptin levels. Cardiac mitochondrial respiration efficiency was decreased by the HFHS diet consumption, which led to reduced ATP and increased NAD(P)H mitochondrial levels, which remained prevented by exercise 8 weeks after cessation. Exercise training-induced cardiac adaptations in redox balance, namely increased relative expression of Nrf2 and downstream antioxidant enzymes persist after an eight-week exercise cessation period. CONCLUSIONS Endurance exercise modulated cardiac redox balance and mitochondrial efficiency in female rats fed a HFHS diet. These findings suggest that exercise may elicit cardiac adaptations crucial for its role as a non-pharmacological intervention for individuals at risk of developing NCDs.
Collapse
Affiliation(s)
- Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - João D Martins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Óscar M Rodrigues
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Jelena Stevanovic-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - David Rizo-Roca
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Estela Santos-Alves
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Manoel Rios
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Lina Carvalho
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - António J Moreno
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, School of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| |
Collapse
|
11
|
Liu Y, Hu J, Wang W, Wang Q. MircroRNA-145 Attenuates Cardiac Fibrosis Via Regulating Mitogen-Activated Protein Kinase Kinase Kinase 3. Cardiovasc Drugs Ther 2023; 37:655-665. [PMID: 35416554 DOI: 10.1007/s10557-021-07312-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE This study aimed to explore the effect of microRNA (miR)-145 on cardiac fibrosis in heart failure mice and its target. METHODS Experiments were carried out in mice receiving left coronary artery ligation, transverse aortic constriction (TAC), or angiotensin (Ang) II to trigger heart failure, and in cardiac fibroblasts (CFs) with Ang II-induced fibrosis. RESULTS The miR-145 levels were decreased in the mice hearts of heart failure induced by myocardial infarction (MI), TAC or Ang II infusion, and in the Ang II-treated CFs. The impaired cardiac function was ameliorated by miR-145 agomiR in MI mice. The increased fibrosis and the levels of collagen I, collagen III, and transforming growth factor-beta (TGF-β) in MI mice were inhibited by miR-145 agomiR or miR-145 transgene (TG). The agomiR of miR-145 also attenuated the increases of collagen I, collagen III, and TGF-β in Ang II-treated CFs. Bioinformatics analysis and luciferase reporter assays indicated that mitogen-activated protein kinase kinase kinase 3 (MAP3K3) was a direct target gene of miR-145. MAP3K3 expression was suppressed by MiR-145 in CFs, while the MAP3K3 over-expression reversed the inhibiting effects of miR-145 agomiR on the Ang II-induced increases of collagen I, collagen III, and TGF-β in CFs. CONCLUSION These results indicated that miR-145 upregulation could improve cardiac dysfunction and cardiac fibrosis by inhibiting MAP3K3 in heart failure. Thus, upregulating miR-145 or blocking MAP3K3 can be used to treat heart failure and cardiac fibrosis.
Collapse
Affiliation(s)
- Yun Liu
- Department of Intensive Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Hu
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Wang
- Department of Intensive Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Pediatric Department, Shanghai General Hospital, No.650 Xinsongjiang Road, Shanghai, 201600, Songjiang District, China.
| |
Collapse
|
12
|
Patel L, Worch JC, Dove AP, Gehmlich K. The Utilisation of Hydrogels for iPSC-Cardiomyocyte Research. Int J Mol Sci 2023; 24:9995. [PMID: 37373141 PMCID: PMC10298477 DOI: 10.3390/ijms24129995] [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: 05/22/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiac fibroblasts' (FBs) and cardiomyocytes' (CMs) behaviour and morphology are influenced by their environment such as remodelling of the myocardium, thus highlighting the importance of biomaterial substrates in cell culture. Biomaterials have emerged as important tools for the development of physiological models, due to the range of adaptable properties of these materials, such as degradability and biocompatibility. Biomaterial hydrogels can act as alternative substrates for cellular studies, which have been particularly key to the progression of the cardiovascular field. This review will focus on the role of hydrogels in cardiac research, specifically the use of natural and synthetic biomaterials such as hyaluronic acid, polydimethylsiloxane and polyethylene glycol for culturing induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). The ability to fine-tune mechanical properties such as stiffness and the versatility of biomaterials is assessed, alongside applications of hydrogels with iPSC-CMs. Natural hydrogels often display higher biocompatibility with iPSC-CMs but often degrade quicker, whereas synthetic hydrogels can be modified to facilitate cell attachment and decrease degradation rates. iPSC-CM structure and electrophysiology can be assessed on natural and synthetic hydrogels, often resolving issues such as immaturity of iPSC-CMs. Biomaterial hydrogels can thus provide a more physiological model of the cardiac extracellular matrix compared to traditional 2D models, with the cardiac field expansively utilising hydrogels to recapitulate disease conditions such as stiffness, encourage alignment of iPSC-CMs and facilitate further model development such as engineered heart tissues (EHTs).
Collapse
Affiliation(s)
- Leena Patel
- Institute of Cardiovascular Science, University of Birmingham, Birmingham B15 2TT, UK;
| | - Joshua C. Worch
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, UK; (J.C.W.); (A.P.D.)
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, UK; (J.C.W.); (A.P.D.)
| | - Katja Gehmlich
- Institute of Cardiovascular Science, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
13
|
Statzer C, Park JYC, Ewald CY. Extracellular Matrix Dynamics as an Emerging yet Understudied Hallmark of Aging and Longevity. Aging Dis 2023; 14:670-693. [PMID: 37191434 DOI: 10.14336/ad.2022.1116] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/16/2022] [Indexed: 05/17/2023] Open
Abstract
The biomechanical properties of extracellular matrices (ECM) and their consequences for cellular homeostasis have recently emerged as a driver of aging. Here we review the age-dependent deterioration of ECM in the context of our current understanding of the aging processes. We discuss the reciprocal interactions of longevity interventions with ECM remodeling. And the relevance of ECM dynamics captured by the matrisome and the matreotypes associated with health, disease, and longevity. Furthermore, we highlight that many established longevity compounds promote ECM homeostasis. A large body of evidence for the ECM to qualify as a hallmark of aging is emerging, and the data in invertebrates is promising. However, direct experimental proof that activating ECM homeostasis is sufficient to slow aging in mammals is lacking. We conclude that further research is required and anticipate that a conceptual framework for ECM biomechanics and homeostasis will provide new strategies to promote health during aging.
Collapse
Affiliation(s)
- Cyril Statzer
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Ji Young Cecilia Park
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| |
Collapse
|
14
|
The Involvement of Krüppel-like Factors in Cardiovascular Diseases. Life (Basel) 2023; 13:life13020420. [PMID: 36836777 PMCID: PMC9962890 DOI: 10.3390/life13020420] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Krüppel-like factors (KLFs) are a set of DNA-binding proteins belonging to a family of zinc-finger transcription factors, which have been associated with many biological processes related to the activation or repression of genes, inducing cell growth, differentiation, and death, and the development and maintenance of tissues. In response to metabolic alterations caused by disease and stress, the heart will undergo cardiac remodeling, leading to cardiovascular diseases (CVDs). KLFs are among the transcriptional factors that take control of many physiological and, in this case, pathophysiological processes of CVD. KLFs seem to be associated with congenital heart disease-linked syndromes, malformations because of autosomal diseases, mutations that relate to protein instability, and/or loss of functions such as atheroprotective activities. Ischemic damage also relates to KLF dysregulation because of the differentiation of cardiac myofibroblasts or a modified fatty acid oxidation related to the formation of a dilated cardiomyopathy, myocardial infarctions, left ventricular hypertrophy, and diabetic cardiomyopathies. In this review, we describe the importance of KLFs in cardiovascular diseases such as atherosclerosis, myocardial infarction, left ventricle hypertrophy, stroke, diabetic cardiomyopathy, and congenital heart diseases. We further discuss microRNAs that have been involved in certain regulatory loops of KLFs as they may act as critical in CVDs.
Collapse
|
15
|
Raza GS, Sodum N, Kaya Y, Herzig KH. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. Int J Mol Sci 2022; 23:12954. [PMID: 36361737 PMCID: PMC9655416 DOI: 10.3390/ijms232112954] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 09/12/2023] Open
Abstract
Circadian rhythms significantly affect metabolism, and their disruption leads to cardiometabolic diseases and fibrosis. The clock repressor Rev-Erb is mainly expressed in the liver, heart, lung, adipose tissue, skeletal muscles, and brain, recognized as a master regulator of metabolism, mitochondrial biogenesis, inflammatory response, and fibrosis. Fibrosis is the response of the body to injuries and chronic inflammation with the accumulation of extracellular matrix in tissues. Activation of myofibroblasts is a key factor in the development of organ fibrosis, initiated by hormones, growth factors, inflammatory cytokines, and mechanical stress. This review summarizes the importance of Rev-Erb in ECM remodeling and tissue fibrosis. In the heart, Rev-Erb activation has been shown to alleviate hypertrophy and increase exercise capacity. In the lung, Rev-Erb agonist reduced pulmonary fibrosis by suppressing fibroblast differentiation. In the liver, Rev-Erb inhibited inflammation and fibrosis by diminishing NF-κB activity. In adipose tissue, Rev- Erb agonists reduced fat mass. In summary, the results of multiple studies in preclinical models demonstrate that Rev-Erb is an attractive target for positively influencing dysregulated metabolism, inflammation, and fibrosis, but more specific tools and studies would be needed to increase the information base for the therapeutic potential of these substances interfering with the molecular clock.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nalini Sodum
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Yagmur Kaya
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Marmara University, 34854 Istanbul, Turkey
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
- Pediatric Gastroenterology and Metabolic Diseases, Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznań, Poland
| |
Collapse
|
16
|
Rusanov VB, Pastushkova LK, Chernikova AG, Kashirina DN, Goncharova AG, Nosovsky AM, Kussmaul AR, Yakhya YD, Popova OV, Brzhozovskiy AG, Orlov OI, Larina IM. Relationship of collagen as the component of the extracellular matrix with the mechanisms of autonomic regulation of the cardiovascular system under simulated conditions of long-term isolation. LIFE SCIENCES IN SPACE RESEARCH 2022; 32:17-25. [PMID: 35065757 DOI: 10.1016/j.lssr.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 06/14/2023]
Abstract
The study of proteins - potential markers, associated signal transduction pathways, and their targets - provides a new understanding of the fundamental mechanisms occurring at the level of regulatory processes in the cardiovascular system (CVS), especially in space flight, as well as in model experiments that reproduce its individual effects on the human body. The article presents the results of studies in an experiment with 120-day isolation within the framework of the SIRIUS project in which 6 volunteers aged 28 to 44 years (three men and three women) participated. SIRIUS (Scientific International Research in Unique Terrestrial Station) is the international research project, which studies the issues of biomedical and psychological support of long-term manned space flights. The possible involvement of collagen different types, an extracellular matrix protein, in the mechanisms of autonomic regulation of the CVS was studied. Using chromatic mass spectrometry in urine samples and analysis of heart rate variability, we have established that the extracellular matrix collagen, which is present, in particular, in the structure of the blood vessel wall, are markers associated with the modulating effect of the autonomic nervous system on the regulatory mechanisms of blood circulation. We hypothesized that these proteins may be a biomarker of the autonomic balance in the regulatory mechanisms of the circulatory system. In addition, these proteins can also be markers of the aging process, which increases the risks of developing autonomic dysfunction of the cardiovascular system (dominance of sympathicotonia) and changes in the quality of the tissue of the heart muscle and blood vessels, provoking the development of prenosological conditions and diseases of the cardiovascular system.
Collapse
Affiliation(s)
- V B Rusanov
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - L Kh Pastushkova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - A G Chernikova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - D N Kashirina
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - A G Goncharova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - A M Nosovsky
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - A R Kussmaul
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation.
| | - Y D Yakhya
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - O V Popova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - A G Brzhozovskiy
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - O I Orlov
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| | - I M Larina
- Institute of Biomedical Problems of the Russian Academy of Sciences, Russian Federation
| |
Collapse
|
17
|
Wang K, Meng X, Guo Z. Elastin Structure, Synthesis, Regulatory Mechanism and Relationship With Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:596702. [PMID: 34917605 PMCID: PMC8670233 DOI: 10.3389/fcell.2021.596702] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/29/2021] [Indexed: 11/30/2022] Open
Abstract
As the primary component of elastic fibers, elastin plays an important role in maintaining the elasticity and tensile ability of cardiovascular, pulmonary and many other tissues and organs. Studies have shown that elastin expression is regulated by a variety of molecules that have positive and negative regulatory effects. However, the specific mechanism is unclear. Moreover, elastin is reportedly involved in the development and progression of many cardiovascular diseases through changes in its expression and structural modifications once deposited in the extracellular matrix. This review article summarizes the role of elastin in myocardial ischemia-reperfusion, atherosclerosis, and atrial fibrillation, with emphasis on the potential molecular regulatory mechanisms.
Collapse
Affiliation(s)
- Keke Wang
- Laboratory of Cardiovascular Disease and Drug Research, Zhengzhou No. 7 People's Hospital, Zhengzhou, China
| | - Xiangguang Meng
- Laboratory of Cardiovascular Disease and Drug Research, Zhengzhou No. 7 People's Hospital, Zhengzhou, China
| | - Zhikun Guo
- Laboratory of Cardiovascular Disease and Drug Research, Zhengzhou No. 7 People's Hospital, Zhengzhou, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
18
|
Wen DT, Zheng L, Lu K, Hou WQ. Physical exercise prevents age-related heart dysfunction induced by high-salt intake and heart salt-specific overexpression in Drosophila. Aging (Albany NY) 2021; 13:19542-19560. [PMID: 34383711 PMCID: PMC8386524 DOI: 10.18632/aging.203364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/17/2021] [Indexed: 12/21/2022]
Abstract
A long-term high-salt intake (HSI) seems to accelerate cardiac aging and age-related diseases, but the molecular mechanism is still not entirely clear. Exercise is an effective way to delay cardiac aging. However, it remains unclear whether long-term exercise (LTE) can protect heart from aging induced by high-salt stress. In this study, heart CG2196(salt) specific overexpression (HSSO) and RNAi (HSSR) was constructed by using the UAS/hand-Gal4 system in Drosophila. Flies were given exercise and a high-salt diet intervention from 1 to 5 weeks of age. Results showed that HSSR and LTE remarkably prevented heart from accelerated age-related defects caused by HSI and HSSO, and these defects included a marked increase in heart period, arrhythmia index, malondialdehyde (MDA) level, salt expression, and dTOR expression, and a marked decrease in fractional shortening, SOD activity level, dFOXO expression, PGC-1α expression, and the number of mitochondria and myofibrils. The combination of HSSR and LTE could better protect the aging heart from the damage of HSI. Therefore, current evidences suggested that LTE resisted HSI-induced heart presenility via blocking CG2196(salt)/TOR/oxidative stress and activating dFOXO/PGC-1α. LTE also reversed heart presenility induced by cardiac-salt overexpression via activating dFOXO/PGC-1α and blocking TOR/oxidative stress.
Collapse
Affiliation(s)
- Deng-Tai Wen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410012, Hunan Province, China.,Ludong University, Yantai 264025, Shandong Province, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410012, Hunan Province, China
| | - Kai Lu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410012, Hunan Province, China
| | - Wen-Qi Hou
- Ludong University, Yantai 264025, Shandong Province, China
| |
Collapse
|
19
|
Lee H, Park J, Park K. Fibrosis as a result of polyhexamethylene guanide exposure in cultured Statens Seruminstitut Rabbit Cornea (SIRC) cells. Environ Anal Health Toxicol 2021; 36:e2021009-0. [PMID: 34130374 PMCID: PMC8421752 DOI: 10.5620/eaht.2021009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
Previous research studies on the toxicity of polyhexamethylene guanidine (PHMG) as a humidifier disinfectant majorly focused on lung fibrosis. Considering that disinfectants in humidifiers are released in aerosol form, the eyes are directly exposed and highly vulnerable to the detrimental effects of the PHMG. Therefore, in the present study we investigated the adverse effects of PHMG on the eyes; considering fibrosis as a manifestation of PHMG toxicity in the eye, we evaluated fibrosis-related biomarkers in cultured Statens Seruminstitut Rabbit Cornea (SIRC) cells. Cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, fibrosis-related biomarkers were evaluated through polymerase chain reaction (PCR) and immunoblotting, and oxidative stress was evaluated using 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA). Polyhexamethylene guanidine showed cytotoxicity in a time and concentration-dependent manner. Fibrosis related biomarkers including transforming growth factor-β (TGF-β), α-smooth muscle actin (α-SMA), matrix metalloproteinase (MMP), tissue inhibitor of metalloproteinase (TIMP) and hemeoxygenase-1 (HO-1) increased in both gene and protein levels. Oxidative stress also increased in the PHMG-treated cultured cells. The findings of the present study suggest that PHMG could cause toxicity in the eye as manifested by fibrosis.
Collapse
Affiliation(s)
- Handule Lee
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Korea
| | - Juyoung Park
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Korea
| | - Kwangsik Park
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Korea
| |
Collapse
|
20
|
Santos ARMP, Jang Y, Son I, Kim J, Park Y. Recapitulating Cardiac Structure and Function In Vitro from Simple to Complex Engineering. MICROMACHINES 2021; 12:mi12040386. [PMID: 33916254 PMCID: PMC8067203 DOI: 10.3390/mi12040386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiac tissue engineering aims to generate in vivo-like functional tissue for the study of cardiac development, homeostasis, and regeneration. Since the heart is composed of various types of cells and extracellular matrix with a specific microenvironment, the fabrication of cardiac tissue in vitro requires integrating technologies of cardiac cells, biomaterials, fabrication, and computational modeling to model the complexity of heart tissue. Here, we review the recent progress of engineering techniques from simple to complex for fabricating matured cardiac tissue in vitro. Advancements in cardiomyocytes, extracellular matrix, geometry, and computational modeling will be discussed based on a technology perspective and their use for preparation of functional cardiac tissue. Since the heart is a very complex system at multiscale levels, an understanding of each technique and their interactions would be highly beneficial to the development of a fully functional heart in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Jongseong Kim
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| | - Yongdoo Park
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| |
Collapse
|
21
|
Szabó R, Hoffmann A, Börzsei D, Kupai K, Veszelka M, Berkó AM, Pávó I, Gesztelyi R, Juhász B, Turcsán Z, Pósa A, Varga C. Hormone Replacement Therapy and Aging: A Potential Therapeutic Approach for Age-Related Oxidative Stress and Cardiac Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8364297. [PMID: 33623635 PMCID: PMC7875635 DOI: 10.1155/2021/8364297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/23/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
Advanced age is an independent risk factor for cardiovascular diseases, which might be further exacerbated by estrogen deficiency. Hormone replacement therapy (HRT) decreases cardiovascular risks and events in postmenopausal women; however, its effects are not fully elucidated in older individuals. Thus, the aim of our study is to examine the impact of HRT on oxidant/antioxidant homeostasis and cardiac remodeling. In our experiment, control (fertile) and aging (~20-month-old) female Wistar rats were used. Aging rats were further divided into estrogen- (E2, 0.1 mg/kg/day per os) or raloxifene- (RAL, 1.0 mg/kg/day per os) treated subgroups. After 2 weeks of treatment, cardiac heme oxygenase (HO) activity, total glutathione (GSH) content, matrix metalloproteinase-2 (MMP-2) activity, and the concentrations of collagen type I and tissue inhibitor of metalloproteinase (TIMP-2), as well as the infarct size, were determined. The aging process significantly decreased the antioxidant HO activity and GSH content, altered the MMP-2/TIMP-2 signaling, and resulted in an excessive collagen accumulation, which culminated in cardiovascular injury. However, 2 weeks of either E2 or RAL treatment enhanced the antioxidant defense mechanisms and attenuated cardiac remodeling related to aging. Our findings clearly show that 2-week-long HRT is a potential intervention to bias successful cardiovascular aging via reducing oxidative damage and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Renáta Szabó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- 1st Department of Medicine, University of Szeged, Szeged H-6720, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Magyariné Berkó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Imre Pávó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Zsolt Turcsán
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Pósa
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
22
|
Effects of Tai Chi Chuan on the Physical and Mental Health of the Elderly: A Systematic Review. PHYSICAL ACTIVITY AND HEALTH 2021. [DOI: 10.5334/paah.70] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
23
|
Cai H, Liu Y, Men H, Zheng Y. Protective Mechanism of Humanin Against Oxidative Stress in Aging-Related Cardiovascular Diseases. Front Endocrinol (Lausanne) 2021; 12:683151. [PMID: 34177809 PMCID: PMC8222669 DOI: 10.3389/fendo.2021.683151] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Physiological reactive oxygen species (ROS) are important regulators of intercellular signal transduction. Oxidative and antioxidation systems maintain a dynamic balance under physiological conditions. Increases in ROS levels destroy the dynamic balance, leading to oxidative stress damage. Oxidative stress is involved in the pathogenesis of aging-related cardiovascular diseases (ACVD), such as atherosclerosis, myocardial infarction, and heart failure, by contributing to apoptosis, hypertrophy, and fibrosis. Oxidative phosphorylation in mitochondria is the main source of ROS. Increasing evidence demonstrates the relationship between ACVD and humanin (HN), an endogenous peptide encoded by mitochondrial DNA. HN protects cardiomyocytes, endothelial cells, and fibroblasts from oxidative stress, highlighting its protective role in atherosclerosis, ischemia-reperfusion injury, and heart failure. Herein, we reviewed the signaling pathways associated with the HN effects on redox signals, including Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2), chaperone-mediated autophagy (CMA), c-jun NH2 terminal kinase (JNK)/p38 mitogen-activated protein kinase (p38 MAPK), adenosine monophosphate-activated protein kinase (AMPK), and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)-Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3). Furthermore, we discussed the relationship among HN, redox signaling pathways, and ACVD. Finally, we propose that HN may be a candidate drug for ACVD.
Collapse
|
24
|
Atale N, Yadav D, Rani V, Jin JO. Pathophysiology, Clinical Characteristics of Diabetic Cardiomyopathy: Therapeutic Potential of Natural Polyphenols. Front Nutr 2020; 7:564352. [PMID: 33344490 PMCID: PMC7744342 DOI: 10.3389/fnut.2020.564352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is an outcome of disturbances in metabolic activities through oxidative stress, local inflammation, and fibrosis, as well as a prime cause of fatality worldwide. Cardiovascular disorders in diabetic individuals have become a challenge in diagnosis and formulation of treatment prototype. It is necessary to have a better understanding of cellular pathophysiology that reveal the therapeutic targets and prevent the progression of cardiovascular diseases due to hyperglycemia. Critical changes in levels of collagen and integrin have been observed in the extracellular matrix of heart, which was responsible for cardiac remodeling in diabetic patients. This review explored the understanding of the mechanisms of how the phytochemicals provide cardioprotection under diabetes along with the caveats and provide future perspectives on these agents as prototypes for the development of drugs for managing DCM. Thus, here we summarized the effect of various plant extracts and natural polyphenols tested in preclinical and cell culture models of diabetic cardiomyopathy. Further, the potential use of selected polyphenols that improved the therapeutic efficacy against diabetic cardiomyopathy is also illustrated.
Collapse
Affiliation(s)
- Neha Atale
- Jaypee Institute of Information Technology, Noida, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Vibha Rani
- Jaypee Institute of Information Technology, Noida, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
25
|
Legumain is a predictor of all-cause mortality and potential therapeutic target in acute myocardial infarction. Cell Death Dis 2020; 11:1014. [PMID: 33243972 PMCID: PMC7691341 DOI: 10.1038/s41419-020-03211-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
The prognostic impact of extracellular matrix (ECM) modulation and its regulatory mechanism post-acute myocardial infarction (AMI), require further clarification. Herein, we explore the predictive role of legumain—which showed the ability in ECM degradation—in an AMI patient cohort and investigate the underlying mechanisms. A total of 212 AMI patients and 323 healthy controls were enrolled in the study. Moreover, AMI was induced in mice by permanent ligation of the left anterior descending artery and fibroblasts were adopted for mechanism analysis. Based on the cut-off value for the receiver-operating characteristics curve, AMI patients were stratified into low (n = 168) and high (n = 44) plasma legumain concentration (PLG) groups. However, PLG was significantly higher in AMI patients than that in the healthy controls (median 5.9 μg/L [interquartile range: 4.2–9.3 μg/L] vs. median 4.4 μg/L [interquartile range: 3.2–6.1 μg/L], P < 0.001). All-cause mortality was significantly higher in the high PLG group compared to that in the low PLG group (median follow-up period, 39.2 months; 31.8% vs. 12.5%; P = 0.002). Multivariate Cox regression analysis showed that high PLG was associated with increased all-cause mortality after adjusting for clinical confounders (HR = 3.1, 95% confidence interval (CI) = 1.4–7.0, P = 0.005). In accordance with the clinical observations, legumain concentration was also increased in peripheral blood, and infarcted cardiac tissue from experimental AMI mice. Pharmacological blockade of legumain with RR-11a, improved cardiac function, decreased cardiac rupture rate, and attenuated left chamber dilation and wall thinning post-AMI. Hence, plasma legumain concentration is of prognostic value in AMI patients. Moreover, legumain aggravates cardiac remodelling through promoting ECM degradation which occurs, at least partially, via activation of the MMP-2 pathway.
Collapse
|
26
|
Choi DH, Oh SY, Choi JK, Lee KE, Lee JY, Park YJ, Jo I, Park YS. A transcriptomic analysis of serial-cultured, tonsil-derived mesenchymal stem cells reveals decreased integrin α3 protein as a potential biomarker of senescent cells. Stem Cell Res Ther 2020; 11:359. [PMID: 32807231 PMCID: PMC7430027 DOI: 10.1186/s13287-020-01860-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been widely used for stem cell therapy, and serial passage of stem cells is often required to obtain sufficient cell numbers for practical applications in regenerative medicine. A long-term serial cell expansion can potentially induce replicative senescence, which leads to a progressive decline in stem cell function and stemness, losing multipotent characteristics. To improve the therapeutic efficiency of stem cell therapy, it would be important to identify specific biomarkers for senescent cells. Methods Tonsil-derived mesenchymal stem cells (TMSCs) with 20–25 passages were designated as culture-aged TMSCs, and their mesodermal differentiation potentials as well as markers of senescence and stemness were compared with the control TMSCs passaged up to 8 times at the most (designated as young). A whole-genome analysis was used to identify novel regulatory factors that distinguish between the culture-aged and control TMSCs. The identified markers of replicative senescence were validated using Western blot analyses. Results The culture-aged TMSCs showed longer doubling time compared to control TMSCs and had higher expression of senescence-associated (SA)-β-gal staining but lower expression of the stemness protein markers, including Nanog, Oct4, and Sox2 with decreased adipogenic, osteogenic, and chondrogenic differentiation potentials. Microarray analyses identified a total of 18,614 differentially expressed genes between the culture-aged and control TMSCs. The differentially expressed genes were classified into the Gene Ontology categories of cellular component (CC), functional component (FC), and biological process (BP) using KEGG (Kyoto encyclopedia of genes and genomes) pathway analysis. This analysis revealed that those genes associated with CC and BP showed the most significant difference between the culture-aged and control TMSCs. The genes related to extracellular matrix-receptor interactions were also shown to be significantly different (p < 0.001). We also found that culture-aged TMSCs had decreased expressions of integrin α3 (ITGA3) and phosphorylated AKT protein (p-AKT-Ser473) compared to the control TMSCs. Conclusions Our data suggest that activation of ECM-receptor signaling, specifically involved with integrin family-mediated activation of the intracellular cell survival-signaling molecule AKT, can regulate stem cell senescence in TMSCs. Among these identified factors, ITGA3 was found to be a representative biomarker of the senescent TMSCs. Exclusion of the TMSCs with the senescent TMSC markers in this study could potentially increase the therapeutic efficacy of TMSCs in clinical applications.
Collapse
Affiliation(s)
- Da Hyeon Choi
- Department of Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Se-Young Oh
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea.,Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - Ju Kwang Choi
- Department of Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Kyeong Eun Lee
- Department of Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Ju Yeon Lee
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Yoon Jeong Park
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.,Department of Dental Regenerative Bioengineering and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea.,Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - Yoon Shin Park
- Department of Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
27
|
Krishnamoorthi MK, Sarig U, Baruch L, Ting S, Reuveny S, Oh S, Goldfracht I, Gepstein L, Venkatraman SS, Tan LP, Machluf M. Robust Fabrication of Composite 3D Scaffolds with Tissue-Specific Bioactivity: A Proof-of-Concept Study. ACS APPLIED BIO MATERIALS 2020; 3:4974-4986. [PMID: 35021675 DOI: 10.1021/acsabm.0c00310] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The basic requirement of any engineered scaffold is to mimic the native tissue extracellular matrix (ECM). Despite substantial strides in understanding the ECM, scaffold fabrication processes of sufficient product robustness and bioactivity require further investigation, owing to the complexity of the natural ECM. A promising bioacive platform for cardiac tissue engineering is that of decellularized porcine cardiac ECM (pcECM, used here as a soft tissue representative model). However, this platform's complexity and batch-to-batch variability serve as processing limitations in attaining a robust and tunable cardiac tissue-specific bioactive scaffold. To address these issues, we fabricated 3D composite scaffolds (3DCSs) that demonstrate comparable physical and biochemical properties to the natural pcECM using wet electrospinning and functionalization with a pcECM hydrogel. The fabricated 3DCSs are non-immunogenic in vitro and support human mesenchymal stem cells' proliferation. Most importantly, the 3DCSs demonstrate tissue-specific bioactivity in inducing spontaneous cardiac lineage differentiation in human induced pluripotent stem cells (hiPSC) and further support the viability, functionality, and maturation of hiPSC-derived cardiomyocytes. Overall, this work illustrates the technology to fabricate robust yet tunable 3D scaffolds of tissue-specific bioactivity (with a proof of concept provided for cardiac tissues) as a platform for basic materials science studies and possible future R&D application in regenerative medicine.
Collapse
Affiliation(s)
- Muthu Kumar Krishnamoorthi
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore.,Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| | - Udi Sarig
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore.,Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel.,Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Guangdong-Technion Israel Institute of Technology (GTIIT), Shantou, Guangdong Province, 515063 P.R. China
| | - Limor Baruch
- Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| | - Sherwin Ting
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, 138668 Singapore
| | - Idit Goldfracht
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Efron St 1, Haifa 31096, Israel
| | - Lior Gepstein
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Efron St 1, Haifa 31096, Israel
| | - Subramanian S Venkatraman
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore
| | - Lay Poh Tan
- School of Materials Science & Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, 639798 Singapore
| | - Marcelle Machluf
- Faculty of Biotechnology & Food Engineering, Technion-Israel Institute of Technology (IIT), Haifa 32000, Israel
| |
Collapse
|
28
|
Koskelainen S, Zhao F, Kalimo H, Baumann M, Kiuru-Enari S. Severe elastolysis in hereditary gelsolin (AGel) amyloidosis. Amyloid 2020; 27:81-88. [PMID: 31814469 DOI: 10.1080/13506129.2019.1699785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AGel amyloidosis is a dominantly inherited systemic amyloidosis caused by mutations p.D214N or p.D214Y resulting in gelsolin amyloid (AGel) formation. AGel accumulates extracellularly in many tissues and alongside elastic fibres. AGel deposition associates with elastic fibre degradation leading to severe clinical manifestations, such as cutis laxa and angiopathic complications. We analysed elastic fibre pathology in dermal and vascular tissue and plasma samples from 35 patients with AGel amyloidosis and 40 control subjects by transmission electron microscopy, immunohistochemistry and ELISA methods. To clarify the pathomechanism(s) of AGel-related elastolysis, we studied the roles of MMP-2, -7, -9, -12 and -14, TIMP-1 and TGFβ. We found massive accumulation of amyloid fibrils along elastic fibres as well as fragmentation and loss of elastic fibres in all dermal and vascular samples of AGel patients. Fibrils of distinct types formed fibrous matrix. The degradation pattern of elastic fibres in AGel patients was different from the age-related degradation in controls. The elastin of elastic fibres in AGel patients was strongly decreased compared to controls. MMP-9 was expressed at lower and TGFβ at higher levels in AGel patients than in controls. The accumulation of amyloid fibrils with severe elastolysis characterises both dermal and vascular derangement in AGel amyloidosis.
Collapse
Affiliation(s)
- Susanna Koskelainen
- Faculty of Medicine, Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland
| | - Fang Zhao
- Faculty of Medicine, Advanced Microscopy Unit, University of Helsinki, Helsinki, Finland
| | - Hannu Kalimo
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marc Baumann
- Faculty of Medicine, Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland
| | - Sari Kiuru-Enari
- Department of Neurosciences, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Saheera S, Krishnamurthy P. Cardiovascular Changes Associated with Hypertensive Heart Disease and Aging. Cell Transplant 2020; 29:963689720920830. [PMID: 32393064 PMCID: PMC7586256 DOI: 10.1177/0963689720920830] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality and morbidity worldwide and account for more than 17.9 million deaths (World Health Organization report). Hypertension and aging are two major risk factors for the development of cardiac structural and functional abnormalities. Hypertension, or elevated blood pressure, if left untreated can result in myocardial hypertrophy leading to heart failure (HF). Left ventricular hypertrophy consequent to pressure overload is recognized as the most important predictor of congestive HF and sudden death. The pathological changes occurring during hypertensive heart disease are very complex and involve many cellular and molecular alterations. In contrast, the cardiac changes that occur with aging are a slow but life-long process and involve all of the structural components in the heart and vasculature. However, these structural changes in the cardiovascular system lead to alterations in overall cardiac physiology and function. The pace at which these pathophysiological changes occur varies between individuals owing to many genetic and environmental risk factors. This review highlights the molecular mechanisms of cardiac structural and functional alterations associated with hypertension and aging.
Collapse
Affiliation(s)
- Sherin Saheera
- Department of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, USA
| |
Collapse
|
30
|
Jiang H, Jia D, Zhang B, Yang W, Dong Z, Sun X, Cui X, Ma L, Wu J, Hu K, Sun A, Ge J. Exercise improves cardiac function and glucose metabolism in mice with experimental myocardial infarction through inhibiting HDAC4 and upregulating GLUT1 expression. Basic Res Cardiol 2020; 115:28. [PMID: 32236769 DOI: 10.1007/s00395-020-0787-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/02/2020] [Indexed: 12/19/2022]
Abstract
This study aims to determine the effect of exercise on the cardiac function, metabolic profiles and related molecular mechanisms in mice with ischemic-induced heart failure (HF). HF was induced by myocardial infarction (MI) in C57BL6/N mice. Cardiac function and physical endurance were improved in HF mice after exercise. Micro-PET/CT scanning revealed enhanced myocardial glucose uptake in vivo in HF mice after exercise. Exercise reduced mitochondrial structural damage in HF mice. Cardiomyocytes isolated from HF + exercise mice showed increased glycolysis capacity, respiratory function and ATP production. Both mRNA and protein expression of glucose transporter 1 (GLUT1) were upregulated after exercise. Results of ChIP-PCR revealed a novel interaction between transcription factor myocyte enhancer factor 2a (MEF2a) and GLUT1 in hearts of HF + exercise mice. Exercise also activated myocardial AMP-activated protein kinase (AMPK), which in turn phosphorylated histone deacetylase 4 (HDAC4), and thereby modulated the GLUT1 expression through reducing its inhibition on MEF2a in HF mice. Inhibition of HDAC4 also improved cardiac function in HF mice. Moreover, knockdown of GLUT1 impaired the systolic and diastolic function of isolated cardiomyocytes. In conclusion, exercise improves cardiac function and glucose metabolism in HF mice through inhibiting HDAC4 and upregulating GLUT1 expression.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Daile Jia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Beijian Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiaotong Cui
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Leilei Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jian Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Institute of Cardiovascular Diseases, Shanghai, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, China. .,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China. .,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Institute of Cardiovascular Diseases, Shanghai, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, China. .,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China. .,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
31
|
Nam JM, Lim JE, Ha TW, Oh B, Kang JO. Cardiac-specific inactivation of Prdm16 effects cardiac conduction abnormalities and cardiomyopathy-associated phenotypes. Am J Physiol Heart Circ Physiol 2020; 318:H764-H777. [PMID: 32083975 DOI: 10.1152/ajpheart.00647.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A variant in the PRDM16 locus has been correlated with QRS duration in an electrocardiogram genome-wide association study, and the deletion of PRDM16 has been implicated as a causal factor of the dilated cardiomyopathy that is linked to 1p36 deletion syndrome. We aimed to determine how a null mutation of Prdm16 affects cardiac function and study the underlying mechanism of the resulting phenotype in an appropriate mouse model. We used cardiac-specific Prdm16 conditional knockout mice to examine cardiac function by electrocardiography. QRS duration and QTc interval increased significantly in cardiac-specific Prdm16 knockout animals compared with wild-type mice. Further, we assessed cardiomyopathy-associated features by trichrome staining, densitometry, and hydroxyproline assay. Prdm16-null hearts showed greater fibrosis and cardiomyocyte hypertrophy. By quantitative real-time PCR, Prdm16-null hearts upregulated extracellular matrix-related genes (Ctgf, Timp1) and α-smooth muscle actin (Acta2), a myofibroblast marker. Moreover, TGF-β signaling was activated in Prdm16-null hearts, as evidenced by increased Tgfb1-3 transcript levels and phosphorylated Smad2. However, the inhibition of TGF-β receptor did not reverse the aberrations in conduction in cardiac-specific Prdm16 knockout mice. To determine the underlying mechanisms, we performed RNA-seq using mouse left ventricular tissue. By functional analysis, Prdm16-null hearts experienced dysregulated expression of ion channel genes, including Kcne1, Scn5a, Cacna1h, and Cacna2d2. Mice with Prdm16-null hearts develop abnormalities in cardiac conduction and cardiomyopathy-associated phenotypes, including fibrosis and cellular hypertrophy. Further, the RNA-seq findings suggest that impairments in ion homeostasis (Ca2+, K+, and Na+) may at least partially underlie the abnormal conduction in cardiac-specific Prdm16 knockout mice.NEW & NOTEWORTHY This is the first study that describes aberrant cardiac function and cardiomyopathy-associated phenotypes in an appropriate murine genetic model with cardiomyocyte-specific Prdm16-null mutation. It is noteworthy that the correlation of PRDM16 with QRS duration is replicated in a murine animal model and the potential underlying mechanism may be the impairment of ion homeostasis.
Collapse
Affiliation(s)
- Jeong Min Nam
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Tae Woong Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
32
|
Szostak J, Wong ET, Titz B, Lee T, Wong SK, Low T, Lee KM, Zhang J, Kumar A, Schlage WK, Guedj E, Phillips B, Leroy P, Buettner A, Xiang Y, Martin F, Sewer A, Kuczaj A, Ivanov NV, Luettich K, Vanscheeuwijck P, Peitsch MC, Hoeng J. A 6-month systems toxicology inhalation study in ApoE -/- mice demonstrates reduced cardiovascular effects of E-vapor aerosols compared with cigarette smoke. Am J Physiol Heart Circ Physiol 2020; 318:H604-H631. [PMID: 31975625 DOI: 10.1152/ajpheart.00613.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Smoking cigarettes is harmful to the cardiovascular system. Considerable attention has been paid to the reduced harm potential of alternative nicotine-containing inhalable products such as e-cigarettes. We investigated the effects of E-vapor aerosols or cigarette smoke (CS) on atherosclerosis progression, cardiovascular function, and molecular changes in the heart and aorta of female apolipoprotein E-deficient (ApoE-/-) mice. The mice were exposed to aerosols from three different E-vapor formulations: 1) carrier (propylene glycol and vegetable glycerol), 2) base (carrier and nicotine), or 3) test (base and flavor) or to CS from 3R4F reference cigarettes for up to 6 mo. Concentrations of CS and base or test aerosols were matched at 35 µg nicotine/L. Exposure to CS, compared with sham-exposed fresh air controls, accelerated atherosclerotic plaque formation, whereas no such effect was seen for any of the three E-vapor aerosols. Molecular changes indicated disease mechanisms related to oxidative stress and inflammation in general, plus changes in calcium regulation, and altered cytoskeletal organization and microtubule dynamics in the left ventricle. While ejection fraction, fractional shortening, cardiac output, and isovolumic contraction time remained unchanged following E-vapor aerosols exposure, the nicotine-containing base and test aerosols caused an increase in isovolumic relaxation time similar to CS. A nicotine-related increase in pulse wave velocity and arterial stiffness was also observed, but it was significantly lower for base and test aerosols than for CS. These results demonstrate that in comparison with CS, E-vapor aerosols induce substantially lower biological responses associated with smoking-related cardiovascular diseases.NEW & NOTEWORTHY Analysis of key urinary oxidative stress markers and proinflammatory cytokines showed an absence of oxidative stress and inflammation in the animals exposed to E-vapor aerosols. Conversely, animals exposed to conventional cigarette smoke had high urinary levels of these markers. When compared with conventional cigarette smoke, E-vapor aerosols induced smaller atherosclerotic plaque surface area and volume. Systolic and diastolic cardiac function, as well as endothelial function, were further significantly less affected by electronic cigarette aerosols than conventional cigarette smoke. Molecular analysis demonstrated that E-vapor aerosols induce significantly smaller transcriptomic dysregulation in the heart and aorta compared with conventional cigarette smoke.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Bjoern Titz
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Tom Lee
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Sin Kei Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Tiffany Low
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | | | | | | | | | - Emmanuel Guedj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Blaine Phillips
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Patrice Leroy
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | | | - Yang Xiang
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Karsta Luettich
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Patrick Vanscheeuwijck
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| |
Collapse
|
33
|
Liu D, Wu L, Wu Y, Wei X, Wang W, Zhang S, Yi M, Li J, Liu H, Ma X. Heat shock factor 1-mediated transcription activation of Omi/HtrA2 induces myocardial mitochondrial apoptosis in the aging heart. Aging (Albany NY) 2019; 11:8982-8997. [PMID: 31627188 PMCID: PMC6834417 DOI: 10.18632/aging.102361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 10/05/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Increased cardiac apoptosis is a hallmark of the elderly, which in turn increases the risk for developing cardiac disease. The overexpression of Omi/HtrA2 mRNA and protein contributes to apoptosis in the aged heart. Heat shock factor 1 (HSF1) is a transcription factor that binds to the promoter of Omi/HtrA2 in the aging myocardium. However, whether HSF1 participates in cardiomyocyte apoptosis via transcriptional regulation of Omi/HtrA2 remains unclear. The present study was designed to investigate whether HSF1 plays a role in Omi/HtrA2 transcriptional regulation and myocardial apoptosis. METHODS AND RESULTS Assessment of the hearts of mice of different ages was performed, which indicated a decrease in cardiac function reserve and an increase in mitochondrial apoptosis. Omi/HtrA2 overexpression in the elderly was negatively correlated with left ventricular function after exercise overload and positively correlated with myocardial Caspase-9 apoptosis. Chromatin immunoprecipitation (ChIP) of aging hearts and plasmid transfection/RNA interference of H9C2 cells revealed that enhancement of HSF1 expression promotes Omi/HtrA2 expression by inducing the promoter activity of Omi/HtrA2 while also increasing mitochondrial apoptosis by upregulating Omi/HtrA2 expression. CONCLUSIONS HSF1 acts as a transcriptional factor that induces Omi/HtrA2 expression and Caspase-9 apoptosis in aged cardiomyocytes, while also decreasing cardiac function reserve.
Collapse
Affiliation(s)
- Dan Liu
- Department of Physiology and Pathophysiology, Yan Jing Medical College, Capital Medical University, Beijing 101300, China
| | - Linguo Wu
- Department of Pathology, Beijing LuHe Hospital of Capital Medical University, Beijing 101100, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xin Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Ming Yi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing Li
- Department of Cardiology, XuanWu Hospital Capital Medical University, Beijing 100053, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
34
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
35
|
Plasma matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs and aging and lifelong exercise adaptations in ventricular and arterial stiffness. Exp Gerontol 2019; 123:36-44. [PMID: 31095969 DOI: 10.1016/j.exger.2019.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 01/19/2023]
Abstract
The age-associated increase in cardiac and central arterial stiffness is attenuated with lifelong (>25 years) endurance exercise in a dose-dependent manner. Remodelling of the extracellular matrix of cardiovascular structures may underpin these lifelong exercise adaptations in structural stiffness. The primary aim was to examine whether matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) levels are associated with aging and lifelong exercise-related changes in cardiac and central arterial stiffness. Plasma MMPs and TIMPs, left ventricular (LV) (LV stiffness constant) and central arterial stiffness (pulse wave velocity) were examined in healthy adults stratified into five groups based on age and lifelong weekly exercise frequency: (1) young sedentary adults (28-50 years), and older adults (>60 years) who had performed either: (a) sedentary (0-1 sessions/week), (b) casual (2-3 sessions/week), (c) committed (4-5 sessions/week) or (d) athletic (≥6 sessions/week) frequency of exercise. MMP-1 was significantly lower in young compared to older sedentary (p = 0.049). Except for TIMP-2 (p = 0.018 versus committed) and the ratio of MMP-2/TIMP-4 (p = 0.047 versus committed), MMP and TIMP expression was not significantly different in lifelong exercise groups (≥casual) compared to the older sedentary group. MMP-1, -3 had a weak positive relationship with central PWV (r = 0.17-0.25, p ≤ 0.050) but there were no significant relationships between MMPs or TIMPs and LV stiffness constant (p ≥ 0.148). In conclusion, there was not a clear or consistent difference in plasma MMPs and TIMPs with lifelong exercise dose despite exhibiting lower cardiovascular stiffness at the highest exercise levels.
Collapse
|
36
|
Gabbai-Armelin PR, Wilian Kido H, Fernandes KR, Fortulan CA, Muniz Renno AC. Effects of bio-inspired bioglass/collagen/magnesium composites on bone repair. J Biomater Appl 2019; 34:261-272. [PMID: 31027447 DOI: 10.1177/0885328219845594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Paulo Roberto Gabbai-Armelin
- 1 Laboratory of Biomaterials and Tissue Engineering, Department of Biosciences, Federal University of São Paulo (UNIFESP), Silva Jardim, Santos, Brazil
| | - Hueliton Wilian Kido
- 1 Laboratory of Biomaterials and Tissue Engineering, Department of Biosciences, Federal University of São Paulo (UNIFESP), Silva Jardim, Santos, Brazil
| | - Kelly Rossetti Fernandes
- 1 Laboratory of Biomaterials and Tissue Engineering, Department of Biosciences, Federal University of São Paulo (UNIFESP), Silva Jardim, Santos, Brazil
| | - Carlos Alberto Fortulan
- 2 Department of Mechanical Engineering, University of São Paulo (USP), Trabalhador São Carlense, São Carlos, Brazil
| | - Ana Claudia Muniz Renno
- 1 Laboratory of Biomaterials and Tissue Engineering, Department of Biosciences, Federal University of São Paulo (UNIFESP), Silva Jardim, Santos, Brazil
| |
Collapse
|
37
|
Cardiac adaptation to exercise training in health and disease. Pflugers Arch 2019; 472:155-168. [PMID: 31016384 DOI: 10.1007/s00424-019-02266-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 02/08/2023]
Abstract
The heart is the primary pump that circulates blood through the entire cardiovascular system, serving many important functions in the body. Exercise training provides favorable anatomical and physiological changes that reduce the risk of heart disease and failure. Compared with pathological cardiac hypertrophy, exercise-induced physiological cardiac hypertrophy leads to an improvement in heart function. Exercise-induced cardiac remodeling is associated with gene regulatory mechanisms and cellular signaling pathways underlying cellular, molecular, and metabolic adaptations. Exercise training also promotes mitochondrial biogenesis and oxidative capacity leading to a decrease in cardiovascular disease. In this review, we summarized the exercise-induced adaptation in cardiac structure and function to understand cellular and molecular signaling pathways and mechanisms in preclinical and clinical trials.
Collapse
|
38
|
Efraim Y, Schoen B, Zahran S, Davidov T, Vasilyev G, Baruch L, Zussman E, Machluf M. 3D Structure and Processing Methods Direct the Biological Attributes of ECM-Based Cardiac Scaffolds. Sci Rep 2019; 9:5578. [PMID: 30944384 PMCID: PMC6447624 DOI: 10.1038/s41598-019-41831-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 03/06/2019] [Indexed: 11/10/2022] Open
Abstract
High hopes are held for cardiac regenerative therapy, driving a vast research effort towards the development of various cardiac scaffolds using diverse technologies and materials. Nevertheless, the role of factors such as fabrication process and structure in determining scaffold's characteristics is yet to be discovered. In the present study, the effects of 3D structure and processing method on cardiac scaffolds are addressed using three distinct scaffolds made through different production technologies from the same biomaterial: decellularized porcine cardiac extracellular matrix (pcECM). pcECM patch, injectable pcECM hydrogel, and electrospun pcECM scaffolds were all proven as viable prospective therapies for MI, thus generally preserving pcECM beneficial properties. Yet, as we demonstrate, minor differences in scaffolds composition and micro-morphology as well as substantial differences in their mechanical properties, which arise from their production process, highly affect the interactions of the scaffold with both proliferating cells and functional cells. Hence, the rates of cell attachment, survival, and proliferation significantly vary between the different scaffolds. Moreover, major differences in cell morphology and alignment as well as in matrix remodeling are obtained. Overall, the effects revealed herein can guide a more rational scaffold design for the improved cellular or acellular treatment of different cardiac disease scenarios.
Collapse
Affiliation(s)
- Yael Efraim
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Beth Schoen
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Sharbel Zahran
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Tzila Davidov
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Gleb Vasilyev
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Limor Baruch
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Eyal Zussman
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Marcelle Machluf
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
39
|
Abstract
The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.
Collapse
Affiliation(s)
- Viktor Wixler
- Centre for Molecular Biology of Inflammation, Institute of Molecular Virology, Westfaelische Wilhelms University Muenster, Muenster, Germany
| |
Collapse
|
40
|
Bejleri D, Davis ME. Decellularized Extracellular Matrix Materials for Cardiac Repair and Regeneration. Adv Healthc Mater 2019; 8:e1801217. [PMID: 30714354 PMCID: PMC7654553 DOI: 10.1002/adhm.201801217] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/20/2018] [Indexed: 12/20/2022]
Abstract
Decellularized extracellular matrix (dECM) is a promising biomaterial for repairing cardiovascular tissue, as dECM most effectively captures the complex array of proteins, glycosaminoglycans, proteoglycans, and many other matrix components that are found in native tissue, providing ideal cues for regeneration and repair of damaged myocardium. dECM can be used in a variety of forms, such as solid scaffolds that maintain native matrix structure, or as soluble materials that can form injectable hydrogels for tissue repair. dECM has found recent success in many regeneration and repair therapies, such as for musculoskeletal, neural, and liver tissues. This review focuses on dECM in the context of cardiovascular applications, with variations in tissue and species sourcing, and specifically discusses advances in solid and soluble dECM development, in vitro studies, in vivo implementation, and clinical translation.
Collapse
Affiliation(s)
- Donald Bejleri
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr., Atlanta, GA, 30322, USA
| | - Michael E Davis
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr., Atlanta, GA, 30322, USA
| |
Collapse
|
41
|
Li S, Liang M, Gao D, Su Q, Laher I. Changes in Titin and Collagen Modulate Effects of Aerobic and Resistance Exercise on Diabetic Cardiac Function. J Cardiovasc Transl Res 2019; 12:404-414. [PMID: 30820865 DOI: 10.1007/s12265-019-09875-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/13/2019] [Indexed: 12/18/2022]
Abstract
Diastolic dysfunction is a common complication that occurs early in diabetes mellitus. Titin and collagen are two important regulators of myocardial passive tension, which contributes to diabetic myocardial diastolic dysfunction. Exercise therapy significantly improves the impaired diabetic cardiac function, but its benefits appear to depend on the type of exercise used. We investigated the effect of aerobic and resistance exercise on cardiac diastolic function in diabetic rats induced by high-fat diet combined with low-dose streptozotocin injection. Interestingly, although resistance training had a more pronounced effect on blood glucose control than did aerobic training in type 2 diabetic rats, improvements in cardiac diastolic parameters benefited more from aerobic training. Moreover, aerobic exercise did significantly increase the expression levels of titin and decrease collagen I, TGFβ1 expression level. In summary, out data suggest that aerobic exercise may improve diabetic cardiac function through changes in titin-dependent myocardial stiffness rather than collagen-dependent interstitial fibrosis.
Collapse
Affiliation(s)
- Shunchang Li
- Institute of Sports Medicine and Health, Chengdu Sport Institute, Chengdu, 610041, China
| | - Min Liang
- Institute of Sports Medicine and Health, Chengdu Sport Institute, Chengdu, 610041, China
| | - Derun Gao
- Institute of Sports Medicine and Health, Chengdu Sport Institute, Chengdu, 610041, China
| | - Quansheng Su
- School of Sports Medicine and Health, Chengdu Sport Institute, Chengdu, 610041, China
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
42
|
Hardy SA, Mabotuwana NS, Murtha LA, Coulter B, Sanchez-Bezanilla S, Al-Omary MS, Senanayake T, Loering S, Starkey M, Lee RJ, Rainer PP, Hansbro PM, Boyle AJ. Novel role of extracellular matrix protein 1 (ECM1) in cardiac aging and myocardial infarction. PLoS One 2019; 14:e0212230. [PMID: 30789914 PMCID: PMC6383988 DOI: 10.1371/journal.pone.0212230] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The prevalence of heart failure increases in the aging population and following myocardial infarction (MI), yet the extracellular matrix (ECM) remodeling underpinning the development of aging- and MI-associated cardiac fibrosis remains poorly understood. A link between inflammation and fibrosis in the heart has long been appreciated, but has mechanistically remained undefined. We investigated the expression of a novel protein, extracellular matrix protein 1 (ECM1) in the aging and infarcted heart. METHODS Young adult (3-month old) and aging (18-month old) C57BL/6 mice were assessed. Young mice were subjected to left anterior descending artery-ligation to induce MI, or transverse aortic constriction (TAC) surgery to induce pressure-overload cardiomyopathy. Left ventricle (LV) tissue was collected early and late post-MI/TAC. Bone marrow cells (BMCs) were isolated from young healthy mice, and subject to flow cytometry. Human cardiac fibroblast (CFb), myocyte, and coronary artery endothelial & smooth muscle cell lines were cultured; human CFbs were treated with recombinant ECM1. Primary mouse CFbs were cultured and treated with recombinant angiotensin-II or TGF-β1. Immunoblotting, qPCR and mRNA fluorescent in-situ hybridization (mRNA-FISH) were conducted on LV tissue and cells. RESULTS ECM1 expression was upregulated in the aging LV, and in the infarct zone of the LV early post-MI. No significant differences in ECM1 expression were found late post-MI or at any time-point post-TAC. ECM1 was not expressed in any resident cardiac cells, but ECM1 was highly expressed in BMCs, with high ECM1 expression in granulocytes. Flow cytometry of bone marrow revealed ECM1 expression in large granular leucocytes. mRNA-FISH revealed that ECM1 was indeed expressed by inflammatory cells in the infarct zone at day-3 post-MI. ECM1 stimulation of CFbs induced ERK1/2 and AKT activation and collagen-I expression, suggesting a pro-fibrotic role. CONCLUSIONS ECM1 expression is increased in ageing and infarcted hearts but is not expressed by resident cardiac cells. Instead it is expressed by bone marrow-derived granulocytes. ECM1 is sufficient to induce cardiac fibroblast stimulation in vitro. Our findings suggest ECM1 is released from infiltrating inflammatory cells, which leads to cardiac fibroblast stimulation and fibrosis in aging and MI. ECM1 may be a novel intermediary between inflammation and fibrosis.
Collapse
Affiliation(s)
- Sean A. Hardy
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nishani S. Mabotuwana
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Lucy A. Murtha
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Brianna Coulter
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Mohammed S. Al-Omary
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Tharindu Senanayake
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Svenja Loering
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Malcolm Starkey
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Randall J. Lee
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
- Edyth and Eli Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States of America
| | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Philip M. Hansbro
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Centre for inflammation, Centenary Institute, Sydney, NSW, Australia
- University of Technology, Faculty of Science, Ultimo, NSW, Australia
| | - Andrew J. Boyle
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
- * E-mail:
| |
Collapse
|
43
|
Parisi JR, Fernandes KR, Avanzi IR, Dorileo BP, Santana AF, Andrade AL, Gabbai-Armelin PR, Fortulan CA, Trichês ES, Granito RN, Renno ACM. Incorporation of Collagen from Marine Sponges (Spongin) into Hydroxyapatite Samples: Characterization and In Vitro Biological Evaluation. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:30-37. [PMID: 30218326 DOI: 10.1007/s10126-018-9855-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/30/2018] [Indexed: 06/08/2023]
Abstract
Biomaterial-based bone grafts have an important role in the field of bone tissue engineering. One of the most promising classes of biomaterials is collagen, including the ones from marine biodiversity (in general, called spongin (SPG)). Also, hydroxyapatite (HA) has an important role in stimulating bone metabolism. Therefore, this work investigated the association of HA and SPG composites in order to evaluate their physico-chemical and morphological characteristics and their in vitro biological performance. For this, pre-set composite disks were evaluated by means of mass loss after incubation, pH, Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and "in vitro" cell viability. pH measurements showed no statistical difference between groups. Moreover, a higher mass loss was observed for HA/SPG70/30 compared to the other groups for all experimental periods. Moreover, SEM representative micrographs showed the degradation of the samples with and without immersion. FTIR analysis demonstrated the absorption peaks for poly(methyl methacrylate) (PMMA), HA, and SPG. A higher L292 cell viability for control and PMMA was observed compared to HA and HA/SPG 90/10. Also, HA/SPG 70/30 showed higher cell viability compared to HA and HA/SPG 90/10 on days 3 and 7 days of culture. Furthermore, HA showed a significant lower MC3T3 cell viability compared to control and HA/SPG 70/30 on day 3 and no significant difference was observed between the composites in the last experimental period. Based on our investigations, it can be concluded that the mentioned composites were successfully obtained, presenting improved biological properties, especially the one mimicking the composition of bone (with 70% of HA and 30% of SPG). Consequently, these data highlight the potential of the introduction of SPG into HA to improve the performance of the graft for bone regeneration applications. Further long-term studies should be carried out to provide additional information concerning the late stages of material degradation and bone healing in the presence of HA/SPG.
Collapse
Affiliation(s)
- J R Parisi
- Department of Physiotherapy, Federal University of São Carlos (UFSCar), Washington Luís, km 235, São Carlos, SP, Brazil.
| | - K R Fernandes
- Department of Physiotherapy, Federal University of São Carlos (UFSCar), Washington Luís, km 235, São Carlos, SP, Brazil
| | - I R Avanzi
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - B P Dorileo
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - A F Santana
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - A L Andrade
- Department of Physiotherapy, Federal University of São Carlos (UFSCar), Washington Luís, km 235, São Carlos, SP, Brazil
| | - P R Gabbai-Armelin
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - C A Fortulan
- Department of Mechanical Engineering, São Carlos School of Engineering São Carlos, São Carlos, SP, Brazil
| | - E S Trichês
- Department of Mechanical Engineering, Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - R N Granito
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - A C M Renno
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| |
Collapse
|
44
|
Abstract
There has been an increasing interest in studying cardiac fibers in order to improve the current knowledge regarding the mechanical and physiological properties of the heart during heart failure (HF), particularly early HF. Having a thorough understanding of the changes in cardiac fiber orientation may provide new insight into the mechanisms behind the progression of left ventricular (LV) remodeling and HF. We conducted a systematic review on various technologies for imaging cardiac fibers and its link to HF. This review covers literature reports from 1900 to 2017. PubMed and Google Scholar databases were searched using the keywords "cardiac fiber" and "heart failure" or "myofiber" and "heart failure." This review highlights imaging methodologies, including magnetic resonance diffusion tensor imaging (MR-DTI), ultrasound, and other imaging technologies as well as their potential applications in basic and translational research on the development and progression of HF. MR-DTI and ultrasound have been most useful and significant in evaluating cardiac fibers and HF. New imaging technologies that have the ability to measure cardiac fiber orientations and identify structural and functional information of the heart will advance basic research and clinical diagnoses of HF.
Collapse
Affiliation(s)
- Shana R Watson
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - James D Dormer
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Baowei Fei
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA. .,Winship Cancer Institute of Emory University, Atlanta, GA, USA. .,Department of Mathematics and Computer Science, Emory University, Atlanta, GA, USA. .,Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA. .,Quantitative Bioimaging Laboratory, Department of Radiology and Imaging Sciences, School of Medicine, Emory University, Atlanta, United States.
| |
Collapse
|
45
|
Toba H, Lindsey ML. Extracellular matrix roles in cardiorenal fibrosis: Potential therapeutic targets for CVD and CKD in the elderly. Pharmacol Ther 2019; 193:99-120. [PMID: 30149103 PMCID: PMC6309764 DOI: 10.1016/j.pharmthera.2018.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Whereas hypertension, diabetes, and dyslipidemia are age-related risk factors for cardiovascular disease (CVD) and chronic kidney disease (CKD), aging alone is an independent risk factor. With advancing age, the heart and kidney gradually but significantly undergo inflammation and subsequent fibrosis, which eventually results in an irreversible decline in organ physiology. Through cardiorenal network interactions, cardiac dysfunction leads to and responds to renal injury, and both facilitate aging effects. Thus, a comprehensive strategy is needed to evaluate the cardiorenal aging network. Common hallmarks shared across systems include extracellular matrix (ECM) accumulation, along with upregulation of matrix metalloproteinases (MMPs) including MMP-9. The wide range of MMP-9 substrates, including ECM components and inflammatory cytokines, implicates MMP-9 in a variety of pathological and age-related processes. In particular, there is strong evidence that inflammatory cell-derived MMP-9 exacerbates cardiorenal aging. This review explores the potential therapeutic targets against CVD and CKD in the elderly, focusing on ECM and MMP roles.
Collapse
Affiliation(s)
- Hiroe Toba
- Department of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
46
|
Effect of Tai Chi on Cardiac and Static Pulmonary Function in Older Community-Dwelling Adults at Risk of Ischemic Stroke: A Randomized Controlled Trial. Chin J Integr Med 2018; 25:582-589. [DOI: 10.1007/s11655-018-3056-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2017] [Indexed: 10/27/2022]
|
47
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
48
|
Wieczfinska J, Kowalczyk T, Sitarek P, Skała E, Pawliczak R. Analysis of Short-Term Smoking Effects in PBMC of Healthy Subjects-Preliminary Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1021. [PMID: 29783661 PMCID: PMC5982060 DOI: 10.3390/ijerph15051021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Abstract
Early structural changes exist in the small airways before the establishment of Chronic Obstructive Pulmonary Disease (COPD). These changes are believed to be induced by oxidation. The aim of this study was to analyze the influence of short-term smoking on the expression of the genes contributing to airway remodeling and their relationship with the oxidative status of human blood cells. Blood mononuclear cells were isolated from 16 healthy volunteers and treated with cigarette smoke ingredients (CSI): nicotine, 1-Nitrosodimethylamine, N-Nitrosopyrrolidyne, vinyl chloride, acetone, and acrolein. The expression of TGF-β1, TIMP-1, SOD1, and arginase I was determined by qPCR. Additionally, thiol groups and TBARs were assessed. CSI induced TGF and TIMP-1 expression in peripheral blood mononuclear cells (PBMC), and apocynin alleviated this effect. The changes were more noticeable in the smoking group (p < 0.05). TBARs concentrations were higher in smokers, and in this group, apocynin acted more effectively. SOD1 correlated with arginase expression in smokers (p < 0.05). MMP-9 showed a significant correlation with SOD1 in both groups, but only on the protein level. Blood cells appear to mirror the general changes caused by cigarette smoke ingredients, which seem to be connected with the oxidative status of the cell. Our findings indicate that a short period of smoking influences the gene expression and oxidative balance of blood cells, which might result in the development of serious disorders such as COPD.
Collapse
Affiliation(s)
- Joanna Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Lodz 90-752, Poland.
| | - Tomasz Kowalczyk
- Department of Genetics and Plant Molecular Biology and Biotechnology, The University of Lodz, Lodz 90-237, Poland.
| | - Przemyslaw Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz 90-151, Poland.
| | - Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz 90-151, Poland.
| | - Rafal Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz 90-752, Poland.
| |
Collapse
|
49
|
Lin YC, Lin YC, Kuo WW, Shen CY, Cheng YC, Lin YM, Chang RL, Padma VV, Huang CY, Huang CY. Platycodin D Reverses Pathological Cardiac Hypertrophy and Fibrosis in Spontaneously Hypertensive Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:537-549. [DOI: 10.1142/s0192415x18500271] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Platycodin D (PD) is the main active saponin isolated from Platycodon grandiflorum (PG) and is reported to exhibit anticancer, anti-angiogenic, anti-inflammation and anti-obesity biological effects. The current study aims to evaluate the therapeutic efficacy of PD in cardiac fibrosis and for hypertrophy in spontaneous hypertension rats (SHRs) and to verify inhibition of the signaling pathway. Significant increases in the cardiac functional indices of left ventricular internal diameter end diastole (LVIDd) and left ventricular internal diameter end systole (LVIDs); the eccentric hypertrophy marker p-MEK5; concentric hypertrophy markers, such as CaMKII[Formula: see text] and calcineurin; and expression levels of NFATc3, p-GATA4 and BNP were observed in spontaneously hypertensive groups. PD treatment reversed these increases in SHRs. In addition, an increase in the fibrosis markers FGF2, uPA, MMP2, MMP9, TGF[Formula: see text]-1 and CTGF during cardiac hypertrophy was detected by western blotting analyses. These results demonstrated that PD treatment considerably attenuates cardiac fibrosis. Histopathological examination revealed that PD treatment remarkably reduced collagen accumulation in contrast to spontaneously hypertensive groups. This study clearly suggests that PD provides myocardial protection by alleviating two damaging responses to hypertension, fibrosis and hypertrophy, in the heart.
Collapse
Affiliation(s)
- Yuan-Chuan Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| | - Yu-Chen Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Chia-Yao Shen
- Department of Nursing, Mei Ho University, Pingtung, Taiwan
| | - Yi-Chang Cheng
- Department of Emergency Medicine, China Medical University, Taichung 40402, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Ruey-Lin Chang
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Vijaya V. Padma
- Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Chih-Yang Huang
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
50
|
Al-Gebaly AS. Ameliorating role of whey syrup against ageing-related damage of myocardial muscle of Wistar Albino rats. Saudi J Biol Sci 2018; 26:950-956. [PMID: 31303824 PMCID: PMC6600591 DOI: 10.1016/j.sjbs.2018.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/23/2018] [Accepted: 03/25/2018] [Indexed: 11/10/2022] Open
Abstract
Age-ing is involved in gradual breakdown of biological structure and function of body organs. The heart represents the main organ responsible for pumping the main issues of life which involving oxygen, nutrients and bioactive molecules necessary for maintaining the body functions. The present study has been conducted to assess the anti-aging properties of whey syrup collected from fermented milk in 4, 18 and 30-months-old rats. The histopathological and histochemical changes of carbohydrates and proteins were investigated. Immunohistochemical expression of smooth muscle actin and P53 was performed to assess the function of cardiomyocytes. Furthermore, Annexin v and biochemical changes of different cardio-biomarkers were carried out to evaluate the effects of aging. The present result of 30 months-old rats revealed myocardial infarction assessed by widening of myocardial fibers, diffused with numerous blood capillaries and dense leukocytic infiltration. The assessed biochemical markers confirmed myocardial damage. Whey supplementation improved the myocardial structure, but less improvement was observed for the 30-months-old rats. The author recommended supplementation with whey is beneficial in giving a body the demand for amino acids and minerals essential for supporting the myocardium and also provides protection against age-ing.
Collapse
Affiliation(s)
- Asma S Al-Gebaly
- Department of Biology, Faculty of Sciences, Princess Nourah Bint Abdulrahman, University, 11474 Riyadh, Saudi Arabia
| |
Collapse
|