1
|
Binlateh T, Hutamekalin P, Yongsawatdigul J, Yamabhai M, Jitprasertwong P. Effects of collagen, chitosan and mixture on fibroblast responses and angiogenic activities in 2D and 3D in vitro models. J Biomed Mater Res A 2023; 111:1642-1655. [PMID: 37222462 DOI: 10.1002/jbm.a.37561] [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: 01/19/2023] [Revised: 04/21/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023]
Abstract
Despite accumulating evidences have demonstrated the potential of collagen and chitosan on tissue repair, it remains unclear on their combination effects. Here, we examined the regenerative effects of single collagen, chitosan and their mixture on fibroblasts and endothelial cells at cellular levels. The results showed that fibroblast responses, as indicated by high proliferative rate, increased spheroid diameter and migrated area existing from spheroid edge, and decreased wound area, were significantly promoted by either collagen or chitosan stimulation. Similarly, both collagen and chitosan resulted in increased endothelial cell proliferation and migration with accelerated tube-like network formation and upregulated VE-cadherin expression, although collagen strongly provided this effect. While the 1:1 mixture (100:100 μg/mL of chitosan to collagen) treatment caused a reduction in fibroblast viability, the lower ratio of chitosan (1:10 mixture; 10:100 μg/mL) did not produce any impact on both fibroblast and endothelial cell viabilities. The 1:10 mixture also significantly enhanced the additional effects on fibroblast responses and angiogenic activities as shown by higher endothelial growth, proliferation and migration with accelerated capillary-like network formation than those treated with the single substance. Further investigation of signaling proteins found that collagen significantly increased expressions of p-Fak, p-Akt and Cdk5 whereas chitosan upregulated p-Fak and Cdk5 expressions. Comparing to the single treatments, p-Fak, p-Akt and Cdk5 were higher expressed in the 1:10 mixture. These observations indicate that proper collagen-chitosan mixture provides the combination effects on fibroblast responses and angiogenic activities when a high concentration of collagen is used, possibly through Fak/Akt and Cdk5 signaling pathways. Therefore, this study helps to define the clinical use of collagen and chitosan as promising biomaterials for tissue repair.
Collapse
Affiliation(s)
- Thunwa Binlateh
- School of Pharmacy, Walailak University, Nakhon Si Thammarat, Thailand
| | - Pilaiwanwadee Hutamekalin
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Jirawat Yongsawatdigul
- Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Montarop Yamabhai
- Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | | |
Collapse
|
2
|
Zhang Y, Yang B, Tu C, Ping Y, Chen S, Wu T, Zhao Z, Mao Y, Yang Z, Cao Z, Li J, Huang K, Ding X, Wu G, Zou P, Deng Z, Sun X. Mitochondrial impairment and downregulation of Drp1 phosphorylation underlie the antiproliferative and proapoptotic effects of alantolactone on oral squamous cell carcinoma cells. J Transl Med 2023; 21:328. [PMID: 37198593 DOI: 10.1186/s12967-023-04188-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most prevalent and fatal oral cancers. Mitochondria-targeting therapies represent promising strategies against various cancers, but their applications in treating OSCC are limited. Alantolactone (ALT) possesses anticancer properties and also regulates mitochondrial events. In this study, we explored the effects of ALT on OSCC and the related mechanisms. METHODS The OSCC cells were treated with varying concentrations and duration of ALT and N-Acetyl-L-cysteine (NAC). The cell viability and colony formation were assessed. The apoptotic rate was evaluated by flow cytometry with Annexin V-FITC/PI double staining. We used DCFH-DA and flow cytometry to detect reactive oxygen species (ROS) production and DAF-FM DA to investigate reactive nitrogen species (RNS) level. Mitochondrial function was reflected by mitochondrial reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and ATP levels. KEGG enrichment analyses determined the mitochondrial-related hub genes involved in OSCC progression. Dynamin-related protein 1 (Drp1) overexpression plasmids were further transfected into the cells to analyze the role of Drp1 in OSCC progression. Immunohistochemistry staining and western blot verified the expression of the protein. RESULTS ALT exerted anti-proliferative and pro-apoptosis effects on OSCC cells. Mechanistically, ALT elicited cell injury by promoting ROS production, mitochondrial membrane depolarization, and ATP depletion, which were reversed by NAC. Bioinformatics analysis showed that Drp1 played a crucial role in OSCC progression. OSCC patients with low Drp1 expression had a higher survival rate. The OSCC cancer tissues presented higher phosphorylated-Drp1 and Drp1 levels than the normal tissues. The results further showed that ALT suppressed Drp1 phosphorylation in OSCC cells. Moreover, Drp1 overexpression abolished the reduced Drp1 phosphorylation by ALT and promoted the cell viability of ALT-treated cells. Drp1 overexpression also reversed the mitochondrial dysfunction induced by ALT, with decreased ROS production, and increased mitochondrial membrane potential and ATP level. CONCLUSIONS ALT inhibited proliferation and promoted apoptosis of oral squamous cell carcinoma cells via impairment of mitochondrial homeostasis and regulation of Drp1. The results provide a solid basis for ALT as a therapeutic candidate for treating OSCC, with Drp1 being a novel therapeutic target in treating OSCC.
Collapse
Affiliation(s)
- Yafei Zhang
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bingqian Yang
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chengwei Tu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam (VU), Amsterdam Movement Sciences (AMS), Amsterdam, The Netherlands
| | - Yifan Ping
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shuhong Chen
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Tong Wu
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zheyu Zhao
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yixin Mao
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhan Yang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zelin Cao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jianmin Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
| | - Kate Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
| | - Xi Ding
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| | - Peng Zou
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Zhennan Deng
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Xiaoyu Sun
- School and Hospital of Stomatology, Institute of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
3
|
Claridge B, Drack A, Pinto AR, Greening DW. Defining cardiac fibrosis complexity and regulation towards therapeutic development. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3. [DOI: 10.1002/ctd2.163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
Abstract
AbstractCardiac fibrosis is insidious, accelerating cardiovascular diseases, heart failure, and death. With a notable lack of effective therapies, advances in both understanding and targeted treatment of fibrosis are urgently needed. Remodelling of the extracellular matrix alters the biomechanical and biochemical cardiac structure and function, disrupting cell‐matrix interactions and exacerbating pathogenesis to ultimately impair cardiac function. Attempts at clinical fibrotic reduction have been fruitless, constrained by an understanding which severely underestimates its dynamic complexity and regulation. Integration of single‐cell sequencing and quantitative proteomics has provided new insights into cardiac fibrosis, including reparative or maladaptive processes, spatiotemporal changes and fibroblast heterogeneity. Further studies have revealed microenvironmental and intercellular signalling mechanisms (including soluble mediators and extracellular vesicles), and intracellular regulators including post‐translational/epigenetic modifications, RNA binding proteins, and non‐coding RNAs. This understanding of novel disease processes and molecular targets has supported the development of innovative therapeutic strategies. Indeed, targeted modulation of cellular heterogeneity, microenvironmental signalling, and intracellular regulation offer promising pre‐clinical therapeutic leads. Clinical development will require further advances in our mechanistic understanding of cardiac fibrosis and dissection of the molecular basis for fibrotic remodelling. This review provides an overview of the complexities of cardiac fibrosis, emerging regulatory mechanisms and therapeutic strategies, and highlights knowledge gaps and opportunities for further investigation towards therapeutic/clinical translation.
Collapse
Affiliation(s)
- Bethany Claridge
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
| | - Auriane Drack
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
| | - Alexander R. Pinto
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
- Baker Department of Cardiometabolic Health University of Melbourne Melbourne Australia
- Central Clinical School Monash University Melbourne Australia
| |
Collapse
|
4
|
Li F, Zhang J, Yi K, Wang H, Wei H, Chan HF, Tao Y, Li M. Delivery of Stem Cell Secretome for Therapeutic Applications. ACS APPLIED BIO MATERIALS 2022; 5:2009-2030. [PMID: 35285638 DOI: 10.1021/acsabm.1c01312] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intensive studies on stem cell therapy reveal that benefits of stem cells attribute to the paracrine effects. Hence, direct delivery of stem cell secretome to the injured site shows the comparative therapeutic efficacy of living cells while avoiding the potential limitations. However, conventional systemic administration of stem cell secretome often leads to rapid clearance in vivo. Therefore, a variety of different biomaterials are developed for sustained and controllable delivery of stem cell secretome to improve therapeutic efficiency. In this review, we first introduce current approaches for the preparation and characterization of stem cell secretome as well as strategies to improve their therapeutic efficacy and production. The up-to-date delivery platforms are also summarized, including nanoparticles, injectable hydrogels, microneedles, and scaffold patches. Meanwhile, we discuss the underlying therapeutic mechanism of stem cell secretome for the treatment of various diseases. In the end, future opportunities and challenges are proposed.
Collapse
Affiliation(s)
- Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hongyan Wei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
5
|
Yamamoto A, Sagara A, Otani K, Okada M, Yamawaki H. Chemerin-9 stimulates migration in rat cardiac fibroblasts in vitro. Eur J Pharmacol 2021; 912:174566. [PMID: 34653380 DOI: 10.1016/j.ejphar.2021.174566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/11/2021] [Indexed: 11/19/2022]
Abstract
Since chemerin is an adipocytokine whose concentration in blood increases in the subjects with various cardiac diseases, chemerin may be involved in pathogenesis of cardiac diseases. In the present study, we examined the effects of chemerin-9, an active fragment of chemerin, on functions of cardiac fibroblasts, which are involved in pathophysiology of cardiac diseases. Primary cardiac fibroblasts were enzymatically isolated from adult male Wistar rats. Migration of cardiac fibroblasts was measured by a Boyden chamber assay and a scratch assay. Phosphorylation of Akt and extracellular signal-regulated kinase (ERK) was measured by Western blotting. Reactive oxygen species (ROS) production was measured by 2',7'-dichlorodihydrofluoresein staining. Chemerin-9 significantly stimulated migration in cardiac fibroblasts. Chemerin-9 significantly stimulated phosphorylation of Akt and ERK as well as ROS production. An Akt pathway inhibitor, LY294002, an ERK pathway inhibitor, PD98059, an antagonist of chemokine-like receptor 1 (CMKLR1), 2-(α-Napththoyl) ethyltrimethylammonium iodide, or an antioxidant, N-acetyl-L-cysteine prevented the migration induced by chemerin-9. In summary, we for the first time revealed that chemerin-9 stimulates migration perhaps through the ROS-dependent activation of Akt and ERK via CMKLR1 in cardiac fibroblasts. It is proposed that chemerin plays a role in the pathogenesis of cardiac diseases.
Collapse
Affiliation(s)
- Atsunori Yamamoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori, 034-8628, Japan
| | - Ayumi Sagara
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori, 034-8628, Japan
| | - Kosuke Otani
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori, 034-8628, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori, 034-8628, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori, 034-8628, Japan.
| |
Collapse
|
6
|
Ambade AS, Hassoun PM, Damico RL. Basement Membrane Extracellular Matrix Proteins in Pulmonary Vascular and Right Ventricular Remodeling in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2021; 65:245-258. [PMID: 34129804 PMCID: PMC8485997 DOI: 10.1165/rcmb.2021-0091tr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM), a highly organized network of structural and nonstructural proteins, plays a pivotal role in cellular and tissue homeostasis. Changes in the ECM are critical for normal tissue repair, whereas dysregulation contributes to aberrant tissue remodeling. Pulmonary arterial hypertension is a severe disorder of the pulmonary vasculature characterized by pathologic remodeling of the pulmonary vasculature and right ventricle, increased production and deposition of structural and nonstructural proteins, and altered expression of ECM growth factors and proteases. Furthermore, ECM remodeling plays a significant role in disease progression, as several dynamic changes in its composition, quantity, and organization are documented in both humans and animal models of disease. These ECM changes impact vascular cell biology and affect proliferation of resident cells. Furthermore, ECM components determine the tissue architecture of the pulmonary and myocardial vasculature as well as the myocardium itself and provide mechanical stability crucial for tissue homeostasis. However, little is known about the basement membrane (BM), a specialized, self-assembled conglomerate of ECM proteins, during remodeling. In the vasculature, the BM is in close physical association with the vascular endothelium and smooth muscle cells. While in the myocardium, each cardiomyocyte is enclosed by a BM that serves as the interface between cardiomyocytes and the surrounding interstitial matrix. In this review, we provide a brief overview on the current state of knowledge of the BM and its ECM composition and their impact on pulmonary vascular remodeling and right ventricle dysfunction and failure in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anjira S Ambade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
7
|
Xu X, Jiang T, Li Y, Kong L. Endostatin attenuates heart failure via inhibiting reactive oxygen species in myocardial infarction rats. Biosci Rep 2021; 41:BSR20200787. [PMID: 32686821 PMCID: PMC8243342 DOI: 10.1042/bsr20200787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/21/2023] Open
Abstract
The purpose of the present study was to evaluate whether endostatin overexpression could improve cardiac function, hemodynamics, and fibrosis in heart failure (HF) via inhibiting reactive oxygen species (ROS). The HF models were established by inducing ischemia myocardial infarction (MI) through ligation of the left anterior descending (LAD) artery in Sprague-Dawley (SD) rats. Endostatin level in serum was increased in MI rats. The decrease in cardiac function and hemodynamics in MI rats were enhanced by endostatin overexpression. Endostatin overexpression inhibited the increase in collagen I, collagen III, α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF), matrix metalloproteinase (MMP)-2 and MMP9 in the hearts of MI rats. MI-induced cardiac hypertrophy was reduced by endostatin overexpression. The increased levels of malondialdehyde (MDA), superoxide anions, the promoted NAD(P)H oxidase (Nox) activity, and the reduced superoxide dismutase (SOD) activity in MI rats were reversed by endostatin overexpression. Nox4 overexpression inhibited the cardiac protective effects of endostatin. These results demonstrated that endostatin improved cardiac dysfunction and hemodynamics, and attenuated cardiac fibrosis and hypertrophy via inhibiting oxidative stress in MI-induced HF rats.
Collapse
Affiliation(s)
- Xuguang Xu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liusha Kong
- Department of Nephrology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
8
|
Frangogiannis NG, Kovacic JC. Extracellular Matrix in Ischemic Heart Disease, Part 4/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2219-2235. [PMID: 32354387 DOI: 10.1016/j.jacc.2020.03.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia and infarction, both in the acute and chronic phases, are associated with cardiomyocyte loss and dramatic changes in the cardiac extracellular matrix (ECM). It has long been appreciated that these changes in the cardiac ECM result in altered mechanical properties of ischemic or infarcted myocardial segments. However, a growing body of evidence now clearly demonstrates that these alterations of the ECM not only affect the structural properties of the ischemic and post-infarct heart, but they also play a crucial and sometimes direct role in mediating a range of biological pathways, including the orchestration of inflammatory and reparative processes, as well as the pathogenesis of adverse remodeling. This final part of a 4-part JACC Focus Seminar reviews the evidence on the role of the ECM in relation to the ischemic and infarcted heart, as well as its contribution to cardiac dysfunction and adverse clinical outcomes.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York.
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
9
|
Pan B, Huo T, Cao M, Jing L, Luo X, Qu Z, Feng H, Yuan F, Guo K. ADAM10 promotes the proliferation of ligamentum flavum cells by activating the PI3K/AKT pathway. Int J Mol Med 2020; 47:688-698. [PMID: 33416124 PMCID: PMC7797459 DOI: 10.3892/ijmm.2020.4809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/30/2020] [Indexed: 01/15/2023] Open
Abstract
Ligamentum flavum hypertrophy (LFH) is an important cause of spinal canal stenosis and posterior longitudinal ligament ossification. Although a number of studies have focused on the mechanisms responsible for LFH, the cellular mechanisms remain poorly understood. The aim of the present study was to investigate the roles of differentially expressed genes (DEGs) in LFH, elucidate the mechanisms responsible for LFH and provide a potential therapeutic target for further studies. The GSE113212 dataset was downloaded from the Gene Expression Omnibus (GEO) database. The microarray data were analyzed and DEGs were obtained. Bioinformatics methods, such as Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and protein-protein interaction (PPI) network analyses were used to obtain the key genes and signaling pathways. In addition, cells derived from hypertrophied ligamentum flavum were cultured, and the key genes and signaling pathways in ligamentum cells were identified through in vitro cell biology and molecular biology experiments. A total of 2,123 genes were screened as DEGs. Among these DEGs, 1,384 genes were upregulated and 739 genes were downregulated. The KEGG pathway analysis revealed that the DEGs were mainly enriched in the PI3K/AKT signaling pathway, and the PPI network analysis screened A disintegrin and metalloproteinase 10 (ADAM10) as a key gene. In vitro experimental verification revealed that ADAM10 promoted the proliferation of ligamentum flavum cells and led to the hypertrophy of the ligamentum by activating the PI3K/AKT pathway. On the whole, the in vitro experimental results suggested that ADAM10 promoted the proliferation of ligamentum flavum cells by activating the PI3K/AKT pathway, which may represent a pathogenic mechanism of LFH. The findings of the present study may provide a basis and direction for further studies on the cellular mechanisms of LFH and present a potential novel therapeutic target and clinical approach.
Collapse
Affiliation(s)
- Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Tianqun Huo
- Department of Orthopedics, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222061, P.R. China
| | - Menghan Cao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Li Jing
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Xuanxiang Luo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Zhe Qu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Hu Feng
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Kaijin Guo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
10
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
11
|
Euscaphic acid and Tormentic acid protect vascular endothelial cells against hypoxia-induced apoptosis via PI3K/AKT or ERK 1/2 signaling pathway. Life Sci 2020; 252:117666. [PMID: 32298737 DOI: 10.1016/j.lfs.2020.117666] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 01/05/2023]
Abstract
AIMS Euscaphic acid and Tormentic acid are aglycones of Kaji-ichigoside F1 and Rosamultin, respectively. These four compounds are pentacyclic triterpenoid, isolated from the subterranean root of the Potentilla anserina L. Based on the protective roles against hypoxia-induced apoptosis of Euscaphic acid and Tormentic acid in vascular endothelial cells, this study was designed to determine the mechanisms. MAIN METHODS The model of hypoxic injuries in EA. hy926 cells was established. Through applications of PI3K/AKT inhibitor, LY294002 and ERK1/2 inhibitor, PD98059, we explored the relationships between pharmacodynamic mechanisms and PI3K/AKT or ERK 1/2 signaling pathway. The anti-hypoxic effects were studied by methyl-thiazolyl-tetrazolium (MTT) assay, Hematoxylin-Eosin (HE) staining, DAPI staining, and flow cytometry. The mechanisms of anti-mitochondrial apoptosis were explored by western blot. The expressions of p-ERK 1/2, ERK 1/2, p-AKT, AKT, p-NF-κB, NF-κB, Bcl-2, Bax, Cyt C, cleaved caspase-9 and cleaved caspase-3 were detected. KEY FINDINGS Euscaphic acid protected vascular endothelial cells against hypoxia-induced apoptosis via ERK1/2 signaling pathway, and Tormentic acid brought its efficacy into full play via PI3K/AKT and ERK1/2 signaling pathways. In addition, PI3K/AKT signaling pathway positively regulated ERK1/2 pathway, and ERK1/2 pathway negatively regulated PI3K/AKT pathway. SIGNIFICANCE This evidence provides theoretical and experimental basis for the following research on anti-hypoxic drugs of Potentilla anserina L.
Collapse
|
12
|
Kaji-Ichigoside F1 and Rosamultin Protect Vascular Endothelial Cells against Hypoxia-Induced Apoptosis via the PI3K/AKT or ERK1/2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6837982. [PMID: 32318240 PMCID: PMC7153006 DOI: 10.1155/2020/6837982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/14/2020] [Accepted: 03/10/2020] [Indexed: 01/23/2023]
Abstract
As a pair of differential isomers, Kaji-ichigoside F1 and Rosamultin are both pentacyclic triterpenoids isolated from the subterranean root of Potentilla anserina L., a plant used in folk medicine in western China as antihypoxia and anti-inflammatory treatments. We demonstrated that Kaji-ichigoside F1 and Rosamultin effectively prevented hypoxia-induced apoptosis in vascular endothelial cells. We established a hypoxia model, using EA.hy926 cells, to further explore the mechanisms. Hypoxia promoted the phosphorylation of AKT, ERK1/2, and NF-κB. In hypoxic cells treated with Kaji-ichigoside F1, p-ERK1/2 and p-NF-κB levels were increased, while the level of p-AKT was decreased. Treatment with Rosamultin promoted phosphorylation of ERK1/2, NF-κB, and AKT in hypoxic cells. Following the addition of LY294002, the levels of p-AKT, p-ERK1/2, and p-NF-κB decreased significantly. Addition of PD98059 resulted in reduced levels of p-ERK1/2 and p-NF-κB, while p-AKT levels were increased. Pharmacodynamic analysis demonstrated that both LY294002 and PD98059 significantly inhibited the positive effects of Kaji-ichigoside F1 on cell viability during hypoxia, consistent with the results of hematoxylin-eosin (H&E) staining, DAPI staining, and flow cytometry. The antihypoxia effects of Rosamultin were remarkably inhibited by LY294002 but promoted by PD98059. In Kaji-ichigoside F1- and Rosamultin-treated cells, Bcl2 expression was significantly upregulated, while expression of Bax and cytochrome C and levels of cleaved caspase-9 and cleaved caspase-3 were reduced. Corresponding to pharmacodynamic analysis, LY294002 inhibited the regulatory effects of Kaji-ichigoside F1 and Rosamultin on the above molecules, while PD98059 inhibited the regulatory effects of Kaji-ichigoside F1 but enhanced the regulatory effects of Rosamultin. In conclusion, Kaji-ichigoside F1 protected vascular endothelial cells against hypoxia-induced apoptosis by activating the ERK1/2 signaling pathway, which positively regulated the NF-κB signaling pathway and negatively regulated the PI3K/AKT signaling pathway. Rosamultin protected vascular endothelial cells against hypoxia-induced apoptosis by activating the PI3K/AKT signaling pathway and positively regulating ERK1/2 and NF-κB signaling pathways.
Collapse
|
13
|
Ushakov A, Ivanchenko V, Gagarina A. Regulation of Myocardial Extracellular Matrix Dynamic Changes in Myocardial Infarction and Postinfarct Remodeling. Curr Cardiol Rev 2020; 16:11-24. [PMID: 31072294 PMCID: PMC7393593 DOI: 10.2174/1573403x15666190509090832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
The article represents literature review dedicated to molecular and cellular mechanisms underlying clinical manifestations and outcomes of acute myocardial infarction. Extracellular matrix adaptive changes are described in detail as one of the most important factors contributing to healing of damaged myocardium and post-infarction cardiac remodeling. Extracellular matrix is reviewed as dynamic constantly remodeling structure that plays a pivotal role in myocardial repair. The role of matrix metalloproteinases and their tissue inhibitors in fragmentation and degradation of extracellular matrix as well as in myocardium healing is discussed. This review provides current information about fibroblasts activity, the role of growth factors, particularly transforming growth factor β and cardiotrophin-1, colony-stimulating factors, adipokines and gastrointestinal hormones, various matricellular proteins. In conclusion considering the fact that dynamic transformation of extracellular matrix after myocardial ischemic damage plays a pivotal role in myocardial infarction outcomes and prognosis, we suggest a high importance of further investigation of mechanisms underlying extracellular matrix remodeling and cell-matrix interactions in cardiovascular diseases.
Collapse
Affiliation(s)
- Alexey Ushakov
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Vera Ivanchenko
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Alina Gagarina
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
14
|
The Non-Fibrillar Side of Fibrosis: Contribution of the Basement Membrane, Proteoglycans, and Glycoproteins to Myocardial Fibrosis. J Cardiovasc Dev Dis 2019; 6:jcdd6040035. [PMID: 31547598 PMCID: PMC6956278 DOI: 10.3390/jcdd6040035] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) provides structural support and a microenvironmentfor soluble extracellular molecules. ECM is comprised of numerous proteins which can be broadly classified as fibrillar (collagen types I and III) and non-fibrillar (basement membrane, proteoglycans, and glycoproteins). The basement membrane provides an interface between the cardiomyocytes and the fibrillar ECM, while proteoglycans sequester soluble growth factors and cytokines. Myocardial fibrosis was originally only linked to accumulation of fibrillar collagens, but is now recognized as the expansion of the ECM including the non-fibrillar ECM proteins. Myocardial fibrosis can be reparative to replace the lost myocardium (e.g., ischemic injury or myocardial infarction), or can be reactive resulting from pathological activity of fibroblasts (e.g., dilated or hypertrophic cardiomyopathy). Contribution of fibrillar collagens to fibrosis is well studied, but the role of the non-fibrillar ECM proteins has remained less explored. In this article, we provide an overview of the contribution of the non-fibrillar components of the extracellular space of the heart to highlight the potential significance of these molecules in fibrosis, with direct evidence for some, although not all of these molecules in their direct contribution to fibrosis.
Collapse
|
15
|
Zhang LL, Du JB, Tang CS, Jin HF, Huang YQ. Inhibitory Effects of Sulfur Dioxide on Rat Myocardial Fibroblast Proliferation and Migration. Chin Med J (Engl) 2018; 131:1715-1723. [PMID: 29998892 PMCID: PMC6048932 DOI: 10.4103/0366-6999.235875] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Myocardial fibrosis is an important pathological change in many heart diseases, but its pathogenesis is very complex and has not yet been fully elucidated. The study was designed to examine whether endogenous sulfur dioxide (SO2) is a novel myocardial fibroblast proliferation and migration inhibitor. Methods: Primary rat myocardial fibroblasts were isolated and transfected with aspartate aminotransferase (AAT1 and AAT2) knockdown lentivirus or empty lentivirus. SO2 content in the supernatant was determined with high-performance liquid chromatography, and the expressions of AAT1, AAT2, proliferating cell nuclear antigen (PCNA), phosphorylated extracellular signal-regulated protein kinase (p-ERK), and total ERK (T-ERK) in the cells were detected. Cell migration was detected by wound healing test. Independent sample t-test (for two groups) and one-way analysis of variance (three or more groups) were used to analyze the results. Results: Both AAT1 and AAT2 knockdown significantly reduced SO2 levels (F = 31.46, P < 0.01) and AAT1/2 protein expression (AAT1, t = 12.67, P < 0.01; AAT2, t = 9.61, P < 0.01), but increased PCNA expression and Cell Counting Kit-8 (CCK-8) activity as well as the migration in rat primary myocardial fibroblasts (P < 0.01). Supplementation of SO2 rather than pyruvate significantly inhibited the increase in proliferation and migration caused by AAT knockdown (P < 0.01). Mechanistically, the ratio of p-ERK to T-ERK was significantly increased in the AAT1/2 knockdown groups compared with that in the empty lentivirus group (AAT1, t = −7.36, P < 0.01; AAT2, t = −10.97, P < 0.01). Whereas PD98059, an inhibitor of ERK activation, successfully blocked AAT knockdown-induced PCNA upregulation (F = 74.01, P > 0.05), CCK-8 activation (F = 50.14, P > 0.05), and migration augmentation in myocardial fibroblasts (24 h, F = 37.08, P > 0.05; 48 h, F = 58.60, P > 0.05). Conclusion: Endogenous SO2 might be a novel myocardial fibroblast proliferation and migration inhibitor via inhibiting the ERK signaling pathway.
Collapse
Affiliation(s)
- Lu-Lu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Jun-Bao Du
- Department of Pediatrics, Peking University First Hospital; Division of Small Molecules and Cardiovascular Disease, Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing 100083, China
| | - Chao-Shu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing 100091, China
| | - Hong-Fang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Ya-Qian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
16
|
Wang C, Luo H, Xu Y, Tao L, Chang C, Shen X. Salvianolic Acid B-Alleviated Angiotensin II Induces Cardiac Fibrosis by Suppressing NF-κB Pathway In Vitro. Med Sci Monit 2018; 24:7654-7664. [PMID: 30365482 PMCID: PMC6215385 DOI: 10.12659/msm.908936] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/22/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Salvianolic acid B (SalB) is the representative component of phenolic acids derived from the roots and rhizomes of Salvia miltiorrhiza Bge (Labiatae), which has been used widely in Asian countries for clinical therapy of various cardiovascular dysfunction-related diseases. However, cardiac protection effects and the underlying mechanism for clinical application are still poorly understood. Here, we investigated the potential anti-myocardial fibrosis effect and mechanism of SalB on Angiotensin II (Ang II)-induced cardiac fibrosis in vitro. MATERIAL AND METHODS The proliferation and migration capacity of cardiac fibroblasts (CFBs) were measured by MTT assay and scratch analysis, respectively. The colorimetric assay determined the hydroxyproline content in medium. Western blotting detected the protein expressions of nuclear transcription factor-kappa B (NF-κB) pathway-associated proteins, fibronectin (FN), collagen type I (Coll I), α-smooth muscle actin (α-SMA), and connective tissue growth factor (CTGF). The expression of α-SMA protein was observed by immunofluorescence staining. qRT-PCR detected the mRNA expression of NF-κB. RESULTS SalB attenuated Ang II-induced the proliferation and the migration ability of CFBs. Ang II-induced the extracellular matrix protein Coll I, FN, and α-SMA, the pro-fibrotic cytokine CTGF protein expression was inhibited, and the nuclear translocation of NF-κB p65 subunit was reduced by SalB. Western blotting and qRT-PCR confirmed that SalB blocked the activation of NF-κB induced by Ang II. PDTC (the NF-κB inhibitor) also inhibited proliferation of CFBs and reduced α-SMA and Coll I expression induced by Ang II. CONCLUSIONS SalB can alleviate Ang II-induced cardiac fibrosis via suppressing the NF-κB pathway in vitro.
Collapse
Affiliation(s)
- Chunhua Wang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| | - Hong Luo
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| | - Yini Xu
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| | - Ling Tao
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| | - Churui Chang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| | - Xiangchun Shen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Drug Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, Guizhou, P.R. China
| |
Collapse
|
17
|
Angelidis A, Račeková E, Arnoul P, Závodská M, Raček A, Martončíková M. Disrupted migration and proliferation of neuroblasts after postnatal administration of angiogenesis inhibitor. Brain Res 2018; 1698:121-129. [PMID: 30092230 DOI: 10.1016/j.brainres.2018.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 02/02/2023]
Abstract
In adult rodents, neuroblasts originating from the subventricular zone migrate tangentially through the rostral migratory stream (RMS) toward the olfactory bulb where they differentiate into interneurons. Neuroblasts in the RMS migrate in chains for a long distance along specifically arranged blood vessels which promote their migration. Although blood vessels in the neurogenic region of the forebrain are present early in development, their rearrangement into this specific pattern takes place during the first postnatal weeks. Here we examined the relevance of this rearrangement to the migration-guiding "scaffold" for the neurogenic processes in the RMS such as cell migration and proliferation. To disturb the reorganization of blood vessels, endostatin - an inhibitor of angiogenesis, was administered systemically to rat pups during the first postnatal week. Ten days or three months later, the arrangement of blood vessels, migration and proliferation of cells in the RMS were assessed. As we expected, the inhibition of angiogenesis disrupted rearrangement of blood vessels in the RMS. The rearrangement's failure resulted in a strong disruption of the mode and direction of neuroblast migration. Chain migration failed and neuroblasts migrated out of the RMS. The inhibition caused a slight increase in the number of proliferating cells in the RMS. The consequences were more obvious ten days after the inhibition of angiogenesis, although they persisted partly into adulthood. Altogether, here we show that the process of rearrangement of blood vessels in the RMS during the early postal period is crucial to ensure the regular course of postnatal neurogenesis.
Collapse
Affiliation(s)
- Andreas Angelidis
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Enikő Račeková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Petra Arnoul
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Monika Závodská
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Adam Raček
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Slovak Republic
| | - Marcela Martončíková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic.
| |
Collapse
|
18
|
Okada M, Imoto K, Sugiyama A, Yasuda J, Yamawaki H. New Insights into the Role of Basement Membrane-Derived Matricryptins in the Heart. Biol Pharm Bull 2018; 40:2050-2060. [PMID: 29199230 DOI: 10.1248/bpb.b17-00308] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM), which contributes to structural homeostasis as well as to the regulation of cellular function, is enzymatically cleaved by proteases, such as matrix metalloproteinases and cathepsins, in the normal and diseased heart. During the past two decades, matricryptins have been defined as fragments of ECM with a biologically active cryptic site, namely the 'matricryptic site,' and their biological activities have been initially identified and clarified, including anti-angiogenic and anti-tumor effects. Thus, matricryptins are expected to be novel anti-tumor drugs, and thus widely investigated. Although there are a smaller number of studies on the expression and function of matricryptins in fields other than cancer research, some matricryptins have been recently clarified to have biological functions beyond an anti-angiogenic effect in heart. This review particularly focuses on the expression and function of basement membrane-derived matricryptins, including arresten, canstatin, tumstatin, endostatin and endorepellin, during cardiac diseases leading to heart failure such as cardiac hypertrophy and myocardial infarction.
Collapse
Affiliation(s)
- Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Keisuke Imoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Akira Sugiyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Jumpei Yasuda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| |
Collapse
|
19
|
Imoto K, Okada M, Yamawaki H. Characterization of fibroblasts from hypertrophied right ventricle of pulmonary hypertensive rats. Pflugers Arch 2018; 470:1405-1417. [PMID: 29860638 DOI: 10.1007/s00424-018-2158-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/07/2018] [Accepted: 05/23/2018] [Indexed: 11/30/2022]
Abstract
Pulmonary arterial hypertension (PAH), which is characterized by an elevation of pulmonary arterial resistance, leads to a lethal right heart failure. It is an urgent issue to clarify the pathogenesis of PAH-induced right heart failure. The present study aimed to elucidate the characteristics of cardiac fibroblasts (CFs) isolated from hypertrophied right ventricles of monocrotaline (MCT)-induced PAH model rats. CFs were isolated from the right ventricles of MCT-injected rats (MCT-CFs) and saline-injected control rats (CONT-CFs). Expression of α-smooth muscle actin and collagen type I in MCT-CFs was lower than that in CONT-CFs. On the other hand, proliferation, migration, and matrix metalloproteinase (MMP)-9 production were significantly enhanced in MCT-CFs. In MCT-CFs, phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, c-Jun N-terminal kinase (JNK), and Ca2+/calmodulin-dependent protein kinase (CaMK) II was significantly enhanced. In addition to mRNA expression of Orai1, a Ca2+ release-activated Ca2+ channel, and stromal interaction molecules (STIM) 1, an endoplasmic reticulum Ca2+ sensor, the associated store-operated Ca2+ entry (SOCE) was significantly higher in MCT-CFs than CONT-CFs. Pharmacological inhibition of ERK1/2 pathway prevented the enhanced proliferation of MCT-CFs. The enhanced migration of MCT-CFs was prevented by a pharmacological inhibition of ERK1/2, JNK, CaMKII, or SOCE pathway. The enhanced MMP-9 production in MCT-CFs was prevented by a pharmacological inhibition of ERK1/2, CaMKII, or SOCE pathway but not JNK. The present results suggested that MCT-CFs exhibit proliferative and migratory phenotypes perhaps through multiple signaling pathways. This study for the first time determined the characteristics of CFs isolated from hypertrophied right ventricles of MCT-induced PAH model rats.
Collapse
Affiliation(s)
- Keisuke Imoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada City, Aomori, 034-8628, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada City, Aomori, 034-8628, Japan.
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada City, Aomori, 034-8628, Japan
| |
Collapse
|
20
|
Waters R, Alam P, Pacelli S, Chakravarti AR, Ahmed RP, Paul A. Stem cell-inspired secretome-rich injectable hydrogel to repair injured cardiac tissue. Acta Biomater 2018; 69:95-106. [PMID: 29281806 DOI: 10.1016/j.actbio.2017.12.025] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/30/2017] [Accepted: 12/18/2017] [Indexed: 12/23/2022]
Abstract
The objective of this study was to develop an injectable and biocompatible hydrogel that can deliver a cocktail of therapeutic biomolecules (secretome) secreted by human adipose-derived stem cells (hASCs) to the peri-infarct myocardium. Gelatin and Laponite® were combined to formulate a shear-thinning, nanocomposite hydrogel (nSi Gel) as an injectable carrier of secretome (nSi Gel+). The growth factor composition and the pro-angiogenic activity of the secretome were tested in vitro by evaluating the proliferation, migration and tube formation of human umbilical endothelial cells. The therapeutic efficacy of the nSi Gel + system was then investigated in vivo in rats by intramyocardial injection into the peri-infarct region. Subsequently, the inflammatory response, angiogenesis, scar formation, and heart function were assessed. Biocompatibility of the developed nSi Gel was confirmed by quantitative PCR and immunohistochemical tests which showed no significant differences in the level of inflammatory genes, microRNAs, and cell marker expression compared to the untreated control group. In addition, the only group that showed a significant increase in capillary density, reduction in scar area and improved cardiac function was treated with the nSi Gel+. Our in vitro and in vivo findings demonstrate the potential of this new secretome-loaded hydrogel as an alternative strategy to treat myocardial infarction. STATEMENT OF SIGNIFICANCE Stem cell based-therapies represent a possible solution to repair damaged myocardial tissue by promoting cardioprotection, angiogenesis, and reduced fibrosis. However, recent evidence indicates that most of the positive outcomes are likely due to the release of paracrine factors (cytokines, growth factors, and exosomes) from the cells and not because of the local engraftment of stem cells. This cocktail of essential growth factors and paracrine signals is known as secretome can be isolated in vitro, and the biomolecule composition can be controlled by varying stem-cell culture conditions. Here, we propose a straightforward strategy to deliver secretome produced from hASCs by using a nanocomposite injectable hydrogel made of gelatin and Laponite®. The designed secretome-loaded hydrogel represents a promising alternative to traditional stem cell therapy for the treatment of acute myocardial infarction.
Collapse
|
21
|
Endostatin Stimulates Proliferation and Migration of Myofibroblasts Isolated from Myocardial Infarction Model Rats. Int J Mol Sci 2018; 19:ijms19030741. [PMID: 29509663 PMCID: PMC5877602 DOI: 10.3390/ijms19030741] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/24/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
Myofibroblasts contribute to the healing of infarcted areas after myocardial infarction through proliferation, migration, and production of extracellular matrix (ECM). Expression of endostatin, a cleaved fragment of type XVIII collagen, increases in the heart tissue of an experimental myocardial infarction model. In the present study, we examined the effect of endostatin on the function of myofibroblasts derived from an infarcted area. The myocardial infarction model was created by ligating the left anterior descending artery in rats. Two weeks after the operation, α-smooth muscle actin (α-SMA)-positive myofibroblasts were isolated from the infarcted area. Endostatin significantly increased the proliferation and migration of myofibroblasts in vitro. On the other hand, endostatin had no effect on the production of type I collagen, a major ECM protein produced by myofibroblasts. Endostatin activated Akt and extracellular signal-regulated kinase (ERK), and the pharmacological inhibition of these signaling pathways suppressed the endostatin-induced proliferation and migration. A knockdown of the COL18A1 gene in the myocardial infarction model rats using small interference RNA (siRNA) worsened the cardiac function concomitant with wall thinning and decreased the α-SMA-positive myofibroblasts and scar formation compared with that of control siRNA-injected rats. In summary, we demonstrated for the first time that endostatin might be an important factor in the healing process after myocardial infarction through the activation of myofibroblasts.
Collapse
|
22
|
Wang L, Yang D, Tian J, Gao A, Shen Y, Ren X, Li X, Jiang G, Dong T. Tumor necrosis factor receptor 2/AKT and ERK signaling pathways contribute to the switch from fibroblasts to CAFs by progranulin in microenvironment of colorectal cancer. Oncotarget 2018; 8:26323-26333. [PMID: 28412748 PMCID: PMC5432260 DOI: 10.18632/oncotarget.15461] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/06/2017] [Indexed: 12/01/2022] Open
Abstract
Cancer associated fibroblasts (CAFs) are a crucial cellular component in tumor microenvironment and could promote tumor progression. CAFs are usually derived from resident fibroblasts, which undergoing an activated process stimulated by tumor cells. However, the agents and mechanism driving this switch have not yet been elucidated. Progranulin (PGRN), a well acknowledged secreted glycoprotein, could promote proliferation and angiogenesis of colorectal cancer (CRC) cells, and high expression of PGRN correlated with patient poor prognosis. Whether PGRN has effects on the function of stromal fibroblasts is unknown. Herein we found that there was a positive correlation between PGRN expression of CRC cells and expressions of smooth muscle actin α (α-SMA) on CAFs in CRC patient tissues. PGRN/α-SMA co-expression was positively correlated with CRC patient poor prognosis. Co-cultured with CRC cells or human recombinant PGRN (rPGRN), the expression of Ki67, fibroblast activation protein (FAP) and α-SMA in fibroblasts were all up-regulated significantly, accompanying with elevated cellular proliferation, migration and contraction. Whilst co-cultured with PGRN-silenced CRC cells, these functions were down-regulated. Studies of the underlying molecular mechanism demonstrated that either tumor necrosis factor receptor 2 (TNFR2)/Akt or the extracellular regulated kinase (ERK) signaling pathway contributed to modulate of Ki67, FAP, and α-SMA expression, and correlated to abilities of proliferation, migration and contraction in fibroblasts. In conclusion, PGRN plays an important role in activation of CRC fibroblasts, which may be taken as a prospective target of CRC therapy.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, Shandong 250117, P. R. China.,Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R.China
| | - Dong Yang
- Department of Oncology, Affiliated hospital of Jining Medical College, Jining, Shandong 272129, P. R. China
| | - Jing Tian
- Department of Oncology, People's Hospital of Zhangqiu City, Zhangqiu, Shandong 250200, P. R. China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R.China
| | - Yihang Shen
- Programs of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI 96813, USA
| | - Xia Ren
- Key Medical Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250012, P. R. China
| | - Xia Li
- Key Medical Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250012, P. R. China
| | - Guosheng Jiang
- Key Medical Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250012, P. R. China
| | - Taotao Dong
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P. R. China
| |
Collapse
|
23
|
Canstatin inhibits isoproterenol-induced apoptosis through preserving mitochondrial morphology in differentiated H9c2 cardiomyoblasts. Apoptosis 2018; 21:887-95. [PMID: 27315818 DOI: 10.1007/s10495-016-1262-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Canstatin, a non-collagenous fragment, is cleaved from type IV collagen α2 chain, an essential component of basement membrane surrounding cardiomyocytes. Although canstatin is known as an endogenous anti-angiogenic factor, its effects on cardiomyocytes have not been clarified. This study examined the effects of canstatin on isoproterenol-induced apoptosis in differentiated H9c2 cardiomyoblasts. Retinoic acid was used to differentiate H9c2 myoblast to cardiomyocyte-like phenotype. Cell viability was determined by a cell counting assay. Western blotting was performed to detect expression of cleaved casepase-3 and phosphorylation of dynamin related protein (Drp)1 at Ser637 which regulates mitochondrial fission. Mito Sox Red staining was performed to examine a mitochondria-dependent production of reactive oxygen species (ROS). Mitochondrial morphology was detected by Mito Tracker Red staining. Isoproterenol (100 μM, 48 h) significantly decreased cell viability and increased cleaved caspase-3 expression, which were inhibited by canstatin (10-250 ng/ml) in a concentration-dependent manner. Canstatin suppressed the isoproterenol-induced mitochondrial fission but not ROS. Canstatin also inhibited the isoproterenol-induced dephosphorylation of Drp1 at Ser637. In conclusion, canstatin inhibits isoproterenol-induced apoptosis through the inhibition of mitochondrial fission via the suppression of dephosphorylation of Drp1 at Ser637 in differentiated H9c2 cardiomyoblasts.
Collapse
|
24
|
T3 peptide, a fragment of tumstatin, stimulates proliferation and migration of cardiac fibroblasts through activation of Akt signaling pathway. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1135-1144. [DOI: 10.1007/s00210-017-1413-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022]
|
25
|
Li X, Li J, Dong X, Gao X, Zhang D, Liu C. A novel 3-Hydroxychromone fluorescence sensor for intracellular Zn2+ and its application in the recognition of prostate cancer cells. SENSORS AND ACTUATORS B: CHEMICAL 2017; 245:129-136. [DOI: 10.1016/j.snb.2017.01.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
|
26
|
Frangogiannis NG. The extracellular matrix in myocardial injury, repair, and remodeling. J Clin Invest 2017; 127:1600-1612. [PMID: 28459429 DOI: 10.1172/jci87491] [Citation(s) in RCA: 356] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cardiac extracellular matrix (ECM) not only provides mechanical support, but also transduces essential molecular signals in health and disease. Following myocardial infarction, dynamic ECM changes drive inflammation and repair. Early generation of bioactive matrix fragments activates proinflammatory signaling. The formation of a highly plastic provisional matrix facilitates leukocyte infiltration and activates infarct myofibroblasts. Deposition of matricellular proteins modulates growth factor signaling and contributes to the spatial and temporal regulation of the reparative response. Mechanical stress due to pressure and volume overload and metabolic dysfunction also induce profound changes in ECM composition that contribute to the pathogenesis of heart failure. This manuscript reviews the role of the ECM in cardiac repair and remodeling and discusses matrix-based therapies that may attenuate remodeling while promoting repair and regeneration.
Collapse
|
27
|
Sugiyama A, Okada M, Yamawaki H. Pathophysiological roles of canstatin on myofibroblasts after myocardial infarction in rats. Eur J Pharmacol 2017; 807:32-43. [PMID: 28438649 DOI: 10.1016/j.ejphar.2017.04.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/13/2017] [Accepted: 04/19/2017] [Indexed: 12/19/2022]
Abstract
Myofibroblasts play an important role during remodeling process after myocardial infarction through proliferation, migration, production and degradation of extracellular matrix (ECM) and contraction. Canstatin, a 24kDa polypeptide, is cleaved from α2 chain of type IV collagen, which is a major component of basement membrane around cardiomyocytes. We examined the effects of canstatin on myofibroblasts isolated from the areas of myocardial infarction. Myocardial infarction model was made by ligating left anterior descending artery of Wistar rats. Two weeks after the operation, the cells were isolated by an explant method and identified as myofibroblasts with immunofluorescence staining. Cell counting assay was performed to examine cell proliferation. Boyden chamber assay was performed to examine cell migration. Expression and phosphorylation of proteins were detected by Western blotting. Collagen gel contraction assay was performed to measure cell contractility. Canstatin stimulated proliferation, secretion of matrix metalloproteinases, expression of cyclooxygenase (COX)-2, and inhibited collagen gel contraction in myofibroblasts. Canstatin increased Akt phosphorylation. LY294002, a phosphoinositide-3-kinase/Akt inhibitor, inhibited the canstatin-induced proliferation. NS-398, a COX-2 inhibitor, suppressed the inhibitory effect of canstatin on collagen gel contraction. Canstatin expression in areas of myocardial infarction 2 weeks after surgery decreased. We for the first time demonstrate that canstatin is an endogenous bioactive molecule regulating the various functions of myofibroblasts after myocardial infarction. The decrease of canstatin expression in the maturated areas of myocardial infarction might lead to stabilization of scar tissues perhaps in part through the reduction of proliferation and ECM degradation as well as the stimulation of contractility in myofibroblasts.
Collapse
Affiliation(s)
- Akira Sugiyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Japan.
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Japan
| |
Collapse
|
28
|
Kanazawa H, Imoto K, Okada M, Yamawaki H. Canstatin inhibits hypoxia-induced apoptosis through activation of integrin/focal adhesion kinase/Akt signaling pathway in H9c2 cardiomyoblasts. PLoS One 2017; 12:e0173051. [PMID: 28235037 PMCID: PMC5325616 DOI: 10.1371/journal.pone.0173051] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/14/2017] [Indexed: 12/19/2022] Open
Abstract
A hypoxic stress which causes apoptosis of cardiomyocytes is the main problem in the ischemic heart disease. Canstatin, a non-collagenous fragment of type IV collagen α2 chain, is an endogenous anti-angiogenic factor. We have previously reported that canstatin has a cytoprotective effect on cardiomyoblasts. In the present study, we examined the effects of canstatin on hypoxia-induced apoptosis in H9c2 cardiomyoblasts. Cell counting assay was performed to determine a cell viability. Western blotting was performed to detect expression of cleaved casepase-3 and phosphorylation of focal adhesion kinase (FAK) and Akt. Immunocytochemical staining was performed to observe a distribution of αv integrin. Hypoxia (1% O2, 48 h) significantly decreased cell viability and increased cleaved caspase-3 expression. Canstatin (10–250 ng/ml) significantly inhibited these changes in a concentration-dependent manner. Cilengitide (1 μM), an αvβ3 and αvβ5 integrin inhibitor, significantly prevented the protective effects of canstatin on cell viability. Canstatin significantly increased phosphorylation of FAK and Akt under hypoxic condition, which were inhibited by cilengitide. LY294002, an inhibitor of phosphatidylinositol-3 kinase/Akt pathway, suppressed the canstatin-induced Akt phosphorylation and reversed the protective effects of canstatin. It was observed that hypoxia caused a localization of αv integrin to focal adhesion. In summary, we for the first time clarified that canstatin inhibits hypoxia-induced apoptosis via FAK and Akt pathways through activating integrins in H9c2 cardiomyoblasts.
Collapse
Affiliation(s)
- Hiroki Kanazawa
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Keisuke Imoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
- * E-mail:
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| |
Collapse
|
29
|
Zhang Y, Yuan H, Sun Y, Wang Y, Wang A. The effects of ethanol on angiogenesis after myocardial infarction, and preservation of angiogenesis with rosuvastatin after heavy drinking. Alcohol 2016; 54:27-32. [PMID: 27565753 DOI: 10.1016/j.alcohol.2016.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 03/27/2016] [Accepted: 05/11/2016] [Indexed: 12/17/2022]
Abstract
The cardioprotective effects of moderate alcohol consumption and statins have been known for years. However, heavy or binge drinking confers a high risk of cardiovascular disease. This study aimed to investigate the effects of different levels of alcohol consumption on acute myocardial infarction that was induced experimentally in rats, with a focus on the potential mechanism of angiogenesis and the effects of statins on heavy drinking. The experimental rats were fed low-dose ethanol (0.5 g/kg/day), high-dose ethanol (5 g/kg/day), and high-dose ethanol with rosuvastatin (10 mg/kg/day) during the entire experiment. Acute myocardial infarctions were induced 4 weeks after the beginning of the experiment. We assessed the capillary density in the myocardium via immunohistochemistry and quantified the expression of vascular endothelial growth factor (VEGF) and endostatin via enzyme-linked immunosorbent assay kits on the 4th day after myocardial infarction. The results revealed that low ethanol consumption promoted angiogenesis in association with higher VEGF and lower endostatin. High ethanol intake suppressed angiogenesis with unchanged VEGF and elevated endostatin. Treatment with rosuvastatin preserved angiogenesis following high ethanol intake, with an upregulation of VEGF. This study highlights that low ethanol consumption obviously promotes angiogenesis in myocardial-infarction rats while increasing the expression of VEGF, whereas high ethanol consumption inhibits ischemia-induced angiogenesis. This study also provides evidence that rosuvastatin alleviates the inhibitory effects of heavy drinking on angiogenesis.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yongle Sun
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yong Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Aihong Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China.
| |
Collapse
|
30
|
Shi H, Lin B, Huang Y, Wu J, Zhang H, Lin C, Wang Z, Zhu J, Zhao Y, Fu X, Lou Z, Li X, Xiao J. Basic fibroblast growth factor promotes melanocyte migration via activating PI3K/Akt-Rac1-FAK-JNK and ERK signaling pathways. IUBMB Life 2016; 68:735-47. [DOI: 10.1002/iub.1531] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 06/07/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Hongxue Shi
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Beibei Lin
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Yan Huang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Jiang Wu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Cai Lin
- Wound Healing and Cell Biology Laboratory; Institute of Basic Medical Science, Chinese PLA General Hospital; Beijing China
| | - Zhouguang Wang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Jingjing Zhu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Yingzhen Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory; Institute of Basic Medical Science, Chinese PLA General Hospital; Beijing China
| | - Zhencai Lou
- Department of Otorhinolaryngology; The Affiliated YiWu Hospital, Wenzhou Medical University; Yiwu China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering; Wenzhou Medical University; Wenzhou China
| |
Collapse
|
31
|
Zhang K, He X, Zhou Y, Gao L, Qi Z, Chen J, Gao X. Atorvastatin Ameliorates Radiation-Induced Cardiac Fibrosis in Rats. Radiat Res 2015; 184:611-20. [DOI: 10.1667/rr14075.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
32
|
Akt-dependent Girdin phosphorylation regulates repair processes after acute myocardial infarction. J Mol Cell Cardiol 2015; 88:55-63. [PMID: 26393439 DOI: 10.1016/j.yjmcc.2015.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is a leading cause of death, and cardiac rupture following myocardial infarction leads to extremely poor prognostic feature. A large body of evidence suggests that Akt is involved in several cardiac diseases. We previously reported that Akt-mediated Girdin phosphorylation is essential for angiogenesis and neointima formation. The role of Girdin expression and phosphorylation in myocardial infarction, however, is not understood. Therefore, we employed Girdin-deficient mice and Girdin S1416A knock-in (Girdin(SA/SA)) mice, replacing the Akt phosphorylation site with alanine, to address this question. We found that Girdin was expressed and phosphorylated in cardiac fibroblasts in vitro and that its phosphorylation was crucial for the proliferation and migration of cardiac fibroblasts. In vivo, Girdin was localized in non-cardiomyocyte interstitial cells and phosphorylated in α-smooth muscle actin-positive cells, which are likely to be cardiac myofibroblasts. In an acute myocardial infarction model, Girdin(SA/SA) suppressed the accumulation and proliferation of cardiac myofibroblasts in the infarcted area. Furthermore, lower collagen deposition in Girdin(SA/SA) mice impaired cardiac repair and resulted in increased mortality attributed to cardiac rupture. These findings suggest an important role of Girdin phosphorylation at serine 1416 in cardiac repair after acute myocardial infarction and provide insights into the complex mechanism of cardiac rupture through the Akt/Girdin-mediated regulation of cardiac myofibroblasts.
Collapse
|
33
|
Yasuda J, Takada L, Kajiwara Y, Okada M, Yamawaki H. Endostatin inhibits bradykinin-induced cardiac contraction. J Vet Med Sci 2015; 77:1391-5. [PMID: 26050753 PMCID: PMC4667655 DOI: 10.1292/jvms.15-0200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
.Endogenous fragments of extracellular matrix are known to possess various biological
effects. Levels of endostatin, a fragment of collagen type XVIII, increase in certain
cardiac diseases, such as cardiac hypertrophy and myocardial infarction. However, the
influence of endostatin on cardiac contraction has not been clarified. In the present
study, we investigated the effects of endostatin on bradykinin-induced atrial contraction.
Isometric contractile force of mouse isolated left atria induced by electrical current
pulse was measured. Voltage-dependent calcium current of guinea pig ventricular myocytes
was measured by a whole-cell patch-clamp technique. Endostatin (100–1,000
ng/ml) alone treatment had no influence on left atrial
contraction. On the other hand, pretreatment with endostatin (300
ng/ml) significantly inhibited bradykinin (1
µM)-induced contraction and voltage-dependent calcium current. These
data suggest that endostatin may decrease bradykinin-induced cardiac contraction perhaps
through the inhibition of voltage-dependent calcium channel.
Collapse
Affiliation(s)
- Jumpei Yasuda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada city, Aomori 034-8628, Japan
| | | | | | | | | |
Collapse
|