1
|
Zhang X, Tian H, Xie C, Yang Y, Li P, Cheng J. The role and mechanism of vascular wall cell ion channels in vascular fibrosis remodeling. Channels (Austin) 2024; 18:2418128. [PMID: 39425532 PMCID: PMC11492694 DOI: 10.1080/19336950.2024.2418128] [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: 06/23/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024] Open
Abstract
Fibrosis is usually the final pathological state of many chronic inflammatory diseases and may lead to organ malfunction. Excessive deposition of extracellular matrix (ECM) molecules is a characteristic of most fibrotic tissues. The blood vessel wall contains three layers of membrane structure, including the intima, which is composed of endothelial cells; the media, which is composed of smooth muscle cells; and the adventitia, which is formed by the interaction of connective tissue and fibroblasts. The occurrence and progression of vascular remodeling are closely associated with cardiovascular diseases, and vascular remodeling can alter the original structure and function of the blood vessel. Dysregulation of the composition of the extracellular matrix in blood vessels leads to the continuous advancement of vascular stiffening and fibrosis. Vascular fibrosis reaction leads to excessive deposition of the extracellular matrix in the vascular adventitia, reduces vessel compliance, and ultimately alters key aspects of vascular biomechanics. The pathogenesis of fibrosis in the vasculature and strategies for its reversal have become interesting and important challenges. Ion channels are widely expressed in the cardiovascular system; they regulate blood pressure, maintain cardiovascular function homeostasis, and play important roles in ion transport, cell differentiation, proliferation. In blood vessels, different types of ion channels in fibroblasts, smooth muscle cells and endothelial cells may be relevant mediators of the development of fibrosis in organs or tissues. This review discusses the known roles of ion channels in vascular fibrosis remodeling and discusses potential therapeutic targets for regulating remodeling and repair after vascular injury.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Hai Tian
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Cheng Xie
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Gonzalez Medina M, Liu Z, Wang J, Zhang C, Cash SB, Cummins CL, Giacca A. Cell-Specific Effects of Insulin in a Murine Model of Restenosis Under Insulin-Sensitive and Insulin-Resistant Conditions. Cells 2024; 13:1387. [PMID: 39195275 DOI: 10.3390/cells13161387] [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: 05/12/2024] [Revised: 07/28/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Restenosis following percutaneous revascularization is a major challenge in patients with insulin resistance and diabetes. Currently, the vascular effects of insulin are not fully understood. In vitro, insulin's effects on endothelial cells (ECs) are beneficial, whereas on vascular smooth muscle cells (SMCs), they are mitogenic. We previously demonstrated a suppressive effect of insulin on neointimal growth under insulin-sensitive conditions that was abolished in insulin-resistant conditions. Here, we aimed to determine the cell-specific effects of insulin on neointimal growth in a model of restenosis under insulin-sensitive and insulin-resistant conditions. Vascular cell-specific insulin receptor (IR)-deficient mice were fed a low-fat diet (LFD) or a high-fat, high-sucrose diet (HFSD) and implanted with an insulin pellet or vehicle prior to femoral artery wire injury. In insulin-sensitive conditions, insulin decreased neointimal growth only in controls. However, under insulin-resistant conditions, insulin had no effect in either control, EC-specific or SMC-specific IR-deficient mice. These data demonstrate that EC and SMC IRs are required for the anti-restenotic effect of insulin in insulin-sensitive conditions and that, in insulin resistance, insulin has no adverse effect on vascular SMCs in vivo.
Collapse
MESH Headings
- Animals
- Insulin Resistance
- Insulin/metabolism
- Insulin/pharmacology
- Mice
- Disease Models, Animal
- Receptor, Insulin/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Neointima/pathology
- Neointima/metabolism
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Marel Gonzalez Medina
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zhiwei Liu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Johny Wang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Cindy Zhang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sarah B Cash
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Adria Giacca
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 3H2, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
3
|
Bickel MA, Sherry DM, Bullen EC, Vance ML, Jones KL, Howard EW, Conley SM. Microvascular smooth muscle cells exhibit divergent phenotypic switching responses to platelet-derived growth factor and insulin-like growth factor 1. Microvasc Res 2024; 151:104609. [PMID: 37716411 PMCID: PMC10842624 DOI: 10.1016/j.mvr.2023.104609] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) phenotypic switching is critical for normal vessel formation, vascular stability, and healthy brain aging. Phenotypic switching is regulated by mediators including platelet derived growth factor (PDGF)-BB, insulin-like growth factor (IGF-1), as well as transforming growth factor-β (TGF-β) and endothelin-1 (ET-1), but much about the role of these factors in microvascular VSMCs remains unclear. METHODS We used primary rat microvascular VSMCs to explore PDGF-BB- and IGF-1-induced phenotypic switching. RESULTS PDGF-BB induced an early proliferative response, followed by formation of polarized leader cells and rapid, directionally coordinated migration. In contrast, IGF-1 induced cell hypertrophy, and only a small degree of migration by unpolarized cells. TGF-β and ET-1 selectively inhibit PDGF-BB-induced VSMC migration primarily by repressing migratory polarization and formation of leader cells. Contractile genes were downregulated by both growth factors, while other genes were differentially regulated by PDGF-BB and IGF-1. CONCLUSIONS These studies indicate that PDGF-BB and IGF-1 stimulate different types of microvascular VSMC phenotypic switching characterized by different modes of cell migration. Our studies are consistent with a chronic vasoprotective role for IGF-1 in VSMCs in the microvasculature while PDGF is more involved in VSMC proliferation and migration in response to acute activities such as neovascularization. Better understanding of the nuances of the phenotypic switching induced by these growth factors is important for our understanding of a variety of microvascular diseases.
Collapse
Affiliation(s)
- Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Ken L Jones
- Bioinformatic Solutions, LLC, Sheridan, WY 82801, United States of America
| | - Eric W Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America.
| |
Collapse
|
4
|
Wu H, Lu Y, Duan Z, Wu J, Lin M, Wu Y, Han S, Li T, Fan Y, Hu X, Xiao H, Feng J, Lu Z, Kong D, Li S. Nanopore long-read RNA sequencing reveals functional alternative splicing variants in human vascular smooth muscle cells. Commun Biol 2023; 6:1104. [PMID: 37907652 PMCID: PMC10618188 DOI: 10.1038/s42003-023-05481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 10/18/2023] [Indexed: 11/02/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the major contributor to vascular repair and remodeling, which showed high level of phenotypic plasticity. Abnormalities in VSMC plasticity can lead to multiple cardiovascular diseases, wherein alternative splicing plays important roles. However, alternative splicing variants in VSMC plasticity are not fully understood. Here we systematically characterized the long-read transcriptome and their dysregulation in human aortic smooth muscle cells (HASMCs) by employing the Oxford Nanopore Technologies long-read RNA sequencing in HASMCs that are separately treated with platelet-derived growth factor, transforming growth factor, and hsa-miR-221-3P transfection. Our analysis reveals frequent alternative splicing events and thousands of unannotated transcripts generated from alternative splicing. HASMCs treated with different factors exhibit distinct transcriptional reprogramming modulated by alternative splicing. We also found that unannotated transcripts produce different open reading frames compared to the annotated transcripts. Finally, we experimentally validated the unannotated transcript derived from gene CISD1, namely CISD1-u, which plays a role in the phenotypic switch of HASMCs. Our study characterizes the phenotypic modulation of HASMCs from an insight of long-read transcriptome, which would promote the understanding and the manipulation of HASMC plasticity in cardiovascular diseases.
Collapse
Affiliation(s)
- Hao Wu
- Department of Cardiovascular Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yicheng Lu
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenzhen Duan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingni Wu
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghui Lin
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangjun Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Siyang Han
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tongqi Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqi Fan
- North Cross School Shanghai, Shanghai, China
| | - Xiaoyuan Hu
- H. Milton Stewart School of Industrial and Systems Engineering, College of Engineering, Geogia Institute of Technology, Atlanta, GA, USA
| | - Hongyan Xiao
- Department of Cardiac Surgery, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Jiaxuan Feng
- Department of Vascular Surgery and Intervention Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqian Lu
- Department of Cardiovascular Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Deping Kong
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shengli Li
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Abe JI, Imanishi M, Li S, Zhang A, Ae Ko K, Samanthapudi VSK, Lee LL, Bojorges AP, Gi YJ, Hobbs BP, Deswal A, Herrmann J, Lin SH, Chini EN, Shen YH, Schadler KL, Nguyen THM, Gupte AA, Reyes-Gibby C, Yeung SCJ, Abe RJ, Olmsted-Davis EA, Krishnan S, Dantzer R, Palaskas NL, Cooke JP, Pownall HJ, Yoshimoto M, Fujiwara K, Hamilton DJ, Burks JK, Wang G, Le NT, Kotla S. An ERK5-NRF2 Axis Mediates Senescence-Associated Stemness and Atherosclerosis. Circ Res 2023; 133:25-44. [PMID: 37264926 PMCID: PMC10357365 DOI: 10.1161/circresaha.122.322017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND ERK5 (extracellular signal-regulated kinase 5) is a dual kinase transcription factor containing an N-terminal kinase domain and a C-terminal transcriptional activation domain. Many ERK5 kinase inhibitors have been developed and tested to treat cancer and inflammatory diseases. However, recent data have raised questions about the role of the catalytic activity of ERK5 in proliferation and inflammation. We aimed to investigate how ERK5 reprograms myeloid cells to the proinflammatory senescent phenotype, subsequently leading to atherosclerosis. METHODS A ERK5 S496A (dephosphorylation mimic) knock in (KI) mouse model was generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), and atherosclerosis was characterized by hypercholesterolemia induction. The plaque phenotyping in homozygous ERK5 S496A KI and wild type (WT) mice was studied using imaging mass cytometry. Bone marrow-derived macrophages were isolated from hypercholesterolemic mice and characterized using RNA sequencing and functional in vitro approaches, including senescence, mitochondria reactive oxygen species, and inflammation assays, as well as by metabolic extracellular flux analysis. RESULTS We show that atherosclerosis was inhibited in ERK5 S496A KI mice. Furthermore, ERK5 S496 phosphorylation mediates both senescence-associated secretory phenotype and senescence-associated stemness by upregulating AHR (aryl hydrocarbon receptor) in plaque and bone marrow-derived macrophages isolated from hypercholesterolemic mice. We also discovered that ERK5 S496 phosphorylation could induce NRF2 (NFE2-related factor 2) SUMOylation at a novel K518 site to inhibit NRF2 transcriptional activity without altering ERK5 catalytic activity and mediates oxidized LDL (low-density lipoprotein)-induced senescence-associated secretory phenotype. Specific ERK5 kinase inhibitors (AX15836 and XMD8-92) also inhibited ERK5 S496 phosphorylation, suggesting the involvement of ERK5 S496 phosphorylation in the anti-inflammatory effects of these ERK5 kinase inhibitors. CONCLUSIONS We discovered a novel mechanism by which the macrophage ERK5-NRF2 axis develops a unique senescence-associated secretory phenotype/stemness phenotype by upregulating AHR to engender atherogenesis. The finding of senescence-associated stemness phenotype provides a molecular explanation to resolve the paradox of senescence in proliferative plaque by permitting myeloid cells to escape the senescence-induced cell cycle arrest during atherosclerosis formation.
Collapse
Affiliation(s)
- Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Shengyu Li
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ling-Ling Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian P. Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Keri L. Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thi-Hong-Minh Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anisha A. Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rei J. Abe
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Henry J. Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
- These authors contributed equally to this work
| | - Jared K. Burks
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors were equivalent co-senior authors
| |
Collapse
|
6
|
Hwang AR, Lee HJ, Kim S, Park SH, Woo CH. Inhibition of p90RSK Ameliorates PDGF-BB-Mediated Phenotypic Change of Vascular Smooth Muscle Cell and Subsequent Hyperplasia of Neointima. Int J Mol Sci 2023; 24:ijms24098094. [PMID: 37175802 PMCID: PMC10179136 DOI: 10.3390/ijms24098094] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/25/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Platelet-derived growth factor type BB (PDGF-BB) regulates vascular smooth muscle cell (VSMC) migration and proliferation, which play critical roles in the development of vascular conditions. p90 ribosomal S6 kinase (p90RSK) can regulate various cellular processes through many different target substrates in several cell types, but the regulatory function of p90RSK on PDGF-BB-mediated cell migration and proliferation and subsequent vascular neointima formation has not yet been extensively examined. In this study, we investigated whether p90RSK inhibition protects VSMCs against PDGF-BB-induced cellular phenotypic changes and the molecular mechanisms underlying the effect of p90RSK inhibition on neointimal hyperplasia in vivo. Pretreatment of cultured primary rat VSMCs with FMK or BI-D1870, which are specific inhibitors of p90RSK, suppressed PDGF-BB-induced phenotypic changes, including migration, proliferation, and extracellular matrix accumulation, in VSMCs. Additionally, FMK and BI-D1870 repressed the PDGF-BB-induced upregulation of cyclin D1 and cyclin-dependent kinase-4 expression. Furthermore, p90RSK inhibition hindered the inhibitory effect of PDGF-BB on Cdk inhibitor p27 expression, indicating that p90RSK may induce VSMC proliferation by regulating the G0/G1 phase. Notably, treatment with FMK resulted in attenuation of neointima development in ligated carotid arteries in mice. The findings imply that p90RSK inhibition mitigates the phenotypic switch and neointimal hyperplasia induced by PDGF-BB.
Collapse
Affiliation(s)
- Ae-Rang Hwang
- Department of Pharmacology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
| | - Hee-Jung Lee
- Department of Pharmacology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
| | - Suji Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeng 2-ro, Osong-eub, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Seong-Hee Park
- Department of Physiology, Ewha Womans University College of Medicine, 25 Magokdong-ro 2-gil, Seoul 07804, Republic of Korea
| | - Chang-Hoon Woo
- Department of Pharmacology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
- Senotherpy-Based Metabolic Disease Control Research Center, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
| |
Collapse
|
7
|
Déglise S, Bechelli C, Allagnat F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front Physiol 2023; 13:1081881. [PMID: 36685215 PMCID: PMC9845604 DOI: 10.3389/fphys.2022.1081881] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Arterial occlusive disease is the leading cause of death in Western countries. Core contemporary therapies for this disease include angioplasties, stents, endarterectomies and bypass surgery. However, these treatments suffer from high failure rates due to re-occlusive vascular wall adaptations and restenosis. Restenosis following vascular surgery is largely due to intimal hyperplasia. Intimal hyperplasia develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel's innermost layer or intima. In this review, we describe the current state of knowledge on the origin and mechanisms underlying the dysregulated proliferation of vascular smooth muscle cells in intimal hyperplasia, and we present the new avenues of research targeting VSMC phenotype and proliferation.
Collapse
Affiliation(s)
| | | | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
8
|
Chen YF, Wu KJ, Siao LR, Tsai HY. Trilinolein, a Natural Triacylglycerol, Protects Cerebral Ischemia through Inhibition of Neuronal Apoptosis and Ameliorates Intimal Hyperplasia via Attenuation of Migration and Modulation of Matrix Metalloproteinase-2 and RAS/MEK/ERK Signaling Pathway in VSMCs. Int J Mol Sci 2022; 23:12820. [PMID: 36361610 PMCID: PMC9658252 DOI: 10.3390/ijms232112820] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/20/2022] [Indexed: 09/11/2023] Open
Abstract
Cerebrovascular disease is one of the leading causes of disability and death worldwide, and seeking a potential treatment is essential. Trilinolein (TriL) is a natural triacylglycerol presented in several plants. The effects of TriL on cerebrovascular diseases such as cerebral ischemia and carotid stenosis have never been studied. Accordingly, we investigated the protection of TriL on cerebral ischemia/reperfusion (I/R) and vascular smooth muscle cell (VSMC) migration in vivo and in vitro. The cerebral infarction area, the intima to media area (I/M ratio), and proliferating cell nuclear antigen (PCNA)-staining of the carotid artery were measured. Platelet-derived growth factor (PDGF)-BB-stimulated A7r5 cell migration and potential mechanisms of TriL were investigated by wound healing, transwell, and Western blotting. TriL (50, 100, and 200 mg/kg, p.o.) reduced: the cerebral infarction area; neurological deficit; TUNEL-positive apoptosis; intimal hyperplasia; and PCNA-positive cells in rodents. TriL (5, 10, and 20 µM) significantly inhibited PDGF-BB-stimulated A7r5 cell migration and reduced matrix metalloproteinase-2 (MMP-2), Ras, MEK, and p-ERK protein levels in PDGF-BB-stimulated A7r5 cells. TriL is protective in models of I/R-induced brain injury, carotid artery ligation-induced intimal hyperplasia, and VSMC migration both in vivo and in vitro. TriL could be potentially efficacious in preventing cerebral ischemia and cerebrovascular diseases.
Collapse
Affiliation(s)
- Yuh-Fung Chen
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
- Department of Pharmacy, China Medical University Hospital, Taichung 404332, Taiwan
| | - Kuo-Jen Wu
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
| | - Lian-Ru Siao
- Department of Pharmacology, China Medical University, Taichung 404333, Taiwan
| | - Huei-Yann Tsai
- Department of Pharmacy, China Medical University Hospital, Taichung 404332, Taiwan
| |
Collapse
|
9
|
An agent-based model of vibration-induced intimal hyperplasia. Biomech Model Mechanobiol 2022; 21:1457-1481. [DOI: 10.1007/s10237-022-01601-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
|
10
|
Yap T, Silickas J, Weerakkody R, Lea T, Santhirakumaran G, Bremner L, Diamantopoulos A, Biasi L, Thomas S, Zayed H, Patel SD. Predictors of outcome in diabetic patients undergoing infrapopliteal endovascular revascularization for chronic limb-threatening ischemia. J Vasc Surg 2021; 75:618-624. [PMID: 34634414 DOI: 10.1016/j.jvs.2021.09.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/22/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The incidence of chronic limb-threatening ischemia in diabetic patients is increasing. The factors influencing outcome after infrapopliteal revascularization in these patients are largely unknown. Therefore, this study aims to identify the impact of perioperative glucose control on the long-term outcomes in this patient cohort, and furthermore to identify other factors independently associated with outcome. METHODS Consecutive diabetic patients undergoing infrapopliteal endovascular revascularization for chronic limb-threatening ischemia were identified. Patients' demographics, procedural details, daily capillary blood glucose, and hemoglobin A1C levels were collected and analyzed against the study end points using Kaplan-Meier and Cox regression analysis. RESULTS A total of 437 infrapopliteal target vessels were successfully crossed in 203 patients. Amputation-free survival by Kaplan-Meier (estimate (standard error)%) was 74 (3.3)% and 63 (3.7)%, primary patency was 61 (4.2)% and 50 (4.9)%, assisted primary patency was 69 (5.2)% and 55 (6.1)%, and secondary patency was 71 (3.8)% and 59 (4.1)% at 1 year and 2 years, respectively. Cox regression analysis showed high perioperative capillary blood glucose levels to be an independent predictor of binary restenosis (hazard ratio [HR], 1.49; 95% confidence interval [CI], 1.31-1.1.78; P = .015). Postprocedural dual-antiplatelet therapy was found to be an independent predictor of amputation-free survival (HR, 1.69; 95% CI, 1.04-2.75; P = .033), and freedom from major adverse limb events (HR: 1.96; 95% CI, 1.16-3.27; P = .023) and baseline estimated glomerular filtration rate was significantly associated with better amputation-free survival (HR, 0.52; 95% CI, 0.31-0.87; P = .014). CONCLUSIONS Poor perioperative glycemic control is associated with a higher incidence of restenosis after infrapopliteal revascularization in diabetic patients. Dual antiplatelet therapy is associated with better outcomes in this group.
Collapse
Affiliation(s)
- Trixie Yap
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom.
| | - Justinas Silickas
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Ruwan Weerakkody
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Talia Lea
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Gowthanan Santhirakumaran
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Laura Bremner
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Athanasios Diamantopoulos
- Department of Interventional Radiology, Guys' and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Lukla Biasi
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Stephen Thomas
- Department of Endocrinology and Metabolic Medicine, Guys' and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Hany Zayed
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Sanjay D Patel
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| |
Collapse
|
11
|
Fu J, Yu MG, Li Q, Park K, King GL. Insulin's actions on vascular tissues: Physiological effects and pathophysiological contributions to vascular complications of diabetes. Mol Metab 2021; 52:101236. [PMID: 33878400 PMCID: PMC8513152 DOI: 10.1016/j.molmet.2021.101236] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background Insulin has been demonstrated to exert direct and indirect effects on vascular tissues. Its actions in vascular cells are mediated by two major pathways: the insulin receptor substrate 1/2-phosphoinositide-3 kinase/Akt (IRS1/2/PI3K/Akt) pathway and the Src/mitogen-activated protein kinase (MAPK) pathway, both of which contribute to the expression and distribution of metabolites, hormones, and cytokines. Scope of review In this review, we summarize the current understanding of insulin's physiological and pathophysiological actions and associated signaling pathways in vascular cells, mainly in endothelial cells (EC) and vascular smooth muscle cells (VSMC), and how these processes lead to selective insulin resistance. We also describe insulin's potential new signaling and biological effects derived from animal studies and cultured capillary and arterial EC, VSMC, and pericytes. We will not provide a detailed discussion of insulin's effects on the myocardium, insulin's structure, or its signaling pathways' various steps, since other articles in this issue discuss these areas in depth. Major conclusions Insulin mediates many important functions on vascular cells via its receptors and signaling cascades. Its direct actions on EC and VSMC are important for transporting and communicating nutrients, cytokines, hormones, and other signaling molecules. These vascular actions are also important for regulating systemic fuel metabolism and energetics. Inhibiting or enhancing these pathways leads to selective insulin resistance, exacerbating the development of endothelial dysfunction, atherosclerosis, restenosis, poor wound healing, and even myocardial dysfunction. Targeted therapies to improve selective insulin resistance in EC and VSMC are thus needed to specifically mitigate these pathological processes. Insulin's actions in vascular cells have a significant influence on systemic metabolism. Insulin exerts its vascular effects through its receptors and signaling cascades. Inhibition or enhancement of different insulin signaling leads to selective insulin resistance. Loss of insulin's actions causes endothelial dysfunction and vascular complications in diabetes.
Collapse
Affiliation(s)
- Jialin Fu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Marc Gregory Yu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Qian Li
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - George L King
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Mittal S, Kamath A, Joseph AM, Rajala MS. PLCγ1‑dependent invasion and migration of cells expressing NSCLC‑associated EGFR mutants. Int J Oncol 2020; 57:989-1000. [PMID: 32945365 DOI: 10.3892/ijo.2020.5112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/11/2020] [Indexed: 11/06/2022] Open
Abstract
The increased tyrosine kinase activity of non‑small cell lung cancer (NSCLC)‑associated epidermal growth factor receptor (EGFR) mutants results in deregulated pathways that contribute to malignant cell survival, tumor progression and metastasis. Previous studies investigating lung cancer‑associated EGFR have focused on the prognostic implications of receptor kinase mutations in patients with NSCLC; however, the role of EGFR mutations in tumor cell invasion and migration remains undetermined. The present study was designed to investigate the role of NSCLC‑associated mutant EGFR‑driven signaling pathways in cell proliferation and invasion. Non‑endogenous EGFR‑expressing 293 cells stably expressing EGFR mutants that are sensitive or resistant to Food and Drug Administration (FDA)‑approved EGFR‑targeted tyrosine kinase inhibitors (TKIs) were used in the present study. The experiments demonstrated an increased phosphorylation of phospholipase (PLC)γ1, c‑Cbl, signal transducer and activator of transcription (Stat), extracellular regulated kinase (Erk)1/2, Akt, Shc and Gab1 proteins in cells expressing a mutant form, rather than the wild‑type receptor. As PLCγ1 is a known regulator of metastatic development, mutant receptor‑mediated PLCγ1 activation was further evaluated. To examine the effects of EGFR and PLCγ1 phosphorylation, the metastatic potential of cells expressing mutants was investigated using wound healing, Transwell cell migration and invasion assays. The inhibition of receptor phosphorylation with the 1st, 2nd and 3rd generation TKIs, gefitinib, afatinib, osimertinib, respectively, reduced PLCγ1 phosphorylation, and reduced the invasive and migratory potential of 293 cells, confirming PLCγ1 as one of the probable downstream effectors of mutant EGFR signaling. However, the PLC inhibitor, U73122, inhibited cell migration and invasion without affecting EGFR signaling and PLCγ1 phosphorylation. Notably, U73122 reduced Akt and Erk1/2 phosphorylation within 25 min of its application; however, 100% cell viability was recorded even after 48 h. Upon further investigation, proliferative signaling pathways remained active at 48 h, in accordance with cell viability. Therefore, the present study concludes that mutant receptor‑mediated PLCγ1 activation may play a significant role in the migration and invasion of NSCLC tumors; however, its regulatory role in tumor cell proliferation warrants further investigation and validation in lung tumor cell lines harboring EGFR mutations.
Collapse
Affiliation(s)
- Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Arpana Kamath
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ann M Joseph
- Department of Biochemistry, Amala Cancer Research Centre, Thrisuur, Kerala 680555, India
| | - Maitreyi S Rajala
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
13
|
Li T, Zhang R, Liu Y, Yao Y, Guo J, Zeng Z. Fufang-Zhenzhu-Tiaozhi capsule ameliorates rabbit's iliac artery restenosis by regulating adiponectin signaling pathway. Biomed Pharmacother 2020; 128:110311. [PMID: 32502838 DOI: 10.1016/j.biopha.2020.110311] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Fufang-Zhenzhu-Tiaozhi Capsule (FTZ), a traditional Chinese medicine, has been shown obvious effects on the treatment of dyslipidemia and atherosclerosis. The aim of this study was to evaluate whether FTZ can ameliorate rabbit iliac artery restenosis after angioplasty by regulating adiponectin signaling pathway. EXPERIMENTAL APPROACH The rabbit iliac artery restenosis model was established through percutaneous iliac artery transluminal balloon angioplasty and a high-fat diet. Twenty eight male New Zealand rabbits (8-week-old) were divided into sham operation group (Group Ⅰ), model group (Group Ⅱ), atorvastatin group (Group Ⅲ) and FTZ group (Group Ⅳ), with 7 rabbits in each group. Vascular stenosis was analyzed with Digital Subtraction Angiography. Level of adiponectin (APN), and inflammatory factor including interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α) as well as monocyte chemoattractant protein-1 (MCP-1) was measured by Enzyme Linked Immunosorbent Assay; and injured iliac artery was collected for Hematoxylin-eosin staining and Western Blotting detection of expression of peroxisome proliferator-activated receptor-alpha (PPAR-α), adenosine 5'-monophosphate -activated protein kinase (AMPK) and phosphorylated adenosine 5'-monophosphate -activated protein kinase (p-AMPK). Besides, we evaluated FTZ's safety for the first time. KEY RESULTS Percutaneous iliac artery transluminal balloon angioplasty and high-fat diet result in inflammatory response and restenosis. Compared with Group Ⅱ, iliac artery restenosis was significantly ameliorated in Group Ⅳ (P < 0.05). Treated with FTZ, serum lipids were significantly decreased (P < 0.01), while the level of APN was elevated significantly (P < 0.01). Western blotting detection of the injured iliac artery showed that the expressions of PPAR-α, AMPK and p-AMPK were significantly increased in Group Ⅳ (P < 0.01) than that in Group Ⅱ. Besides, before and after taking drugs, liver and kidney function indicators, creatine kinase, as well as measurement of echocardiography were of no statistical difference in four groups(P > 0.05). CONCLUSIONS AND IMPLICATIONS FTZ could effectively reduce serum lipids and ameliorate rabbit's iliac artery restenosis after angioplasty, and its mechanism may be related to activation of APN signaling pathway.
Collapse
Affiliation(s)
- Tudi Li
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Rendan Zhang
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Yuhong Liu
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Yusi Yao
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Jiao Guo
- Guangdong Pharmaceutical University, China.
| | - Zhihuan Zeng
- Department of Cardiovascular Diseases, the First Affiliated Hospital of Guangdong Pharmaceutical University, China.
| |
Collapse
|
14
|
Oka M, Shimo S, Ohno N, Imai H, Abekura Y, Koseki H, Miyata H, Shimizu K, Kushamae M, Ono I, Nozaki K, Kawashima A, Kawamata T, Aoki T. Dedifferentiation of smooth muscle cells in intracranial aneurysms and its potential contribution to the pathogenesis. Sci Rep 2020; 10:8330. [PMID: 32433495 PMCID: PMC7239886 DOI: 10.1038/s41598-020-65361-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
Smooth muscle cells (SMCs) are the major type of cells constituting arterial walls and play a role to maintain stiffness via producing extracellular matrix. Here, the loss and degenerative changes of SMCs become the major histopathological features of an intracranial aneurysm (IA), a major cause of subarachnoid hemorrhage. Considering the important role of SMCs and the loss of this type of cells in IA lesions, we in the present study subjected rats to IA models and examined how SMCs behave during disease progression. We found that, at the neck portion of IAs, SMCs accumulated underneath the internal elastic lamina according to disease progression and formed the intimal hyperplasia. As these SMCs were positive for a dedifferentiation marker, myosin heavy chain 10, and contained abundant mitochondria and rough endoplasmic reticulum, SMCs at the intimal hyperplasia were dedifferentiated and activated. Furthermore, dedifferentiated SMCs expressed some pro-inflammatory factors, suggesting the role in the formation of inflammatory microenvironment to promote the disease. Intriguingly, some SMCs at the intimal hyperplasia were positive for CD68 and contained lipid depositions, indicating similarity with atherosclerosis. We next examined a potential factor mediating dedifferentiation and recruitment of SMCs. Platelet derived growth factor (PDGF)-BB was expressed in endothelial cells at the neck portion of lesions where high wall shear stress (WSS) was loaded. PDGF-BB facilitated migration of SMCs across matrigel-coated pores in a transwell system, promoted dedifferentiation of SMCs and induced expression of pro-inflammatory genes in these cells in vitro. Because, in a stenosis model of rats, PDGF-BB expression was expressed in endothelial cells loaded in high WSS regions, and SMCs present nearby were dedifferentiated, hence a correlation existed between high WSS, PDGFB and dedifferentiation in vivo. In conclusion, dedifferentiated SMCs presumably by PDGF-BB produced from high WSS-loaded endothelial cells accumulate in the intimal hyperplasia to form inflammatory microenvironment leading to the progression of the disease.
Collapse
Affiliation(s)
- Mieko Oka
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, 8-1 Kawata-cho, Shinjyuku-ku, Tokyo, 162-8666, Japan
| | - Satoshi Shimo
- Department of Occupational Therapy, Health Science University, 7181 Kodachi, Minamitsurugun Fujikawaguchikomachi, Yamanashi, 401-0380, Japan
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke City, Tochigi, 329-0498, Japan.,Division of Ultrastructural Research, National Institute for Physiological Sciences, 38 Saigonaka, Meidaiji-cho, Okazaki City, Aichi, 444-8787, Japan
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, 36-1 Yoshidahomachi Saikyo-ku, Kyoto City, Kyoto, 606-8317, Japan
| | - Yu Abekura
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hirokazu Koseki
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Haruka Miyata
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Kampei Shimizu
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mika Kushamae
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan
| | - Isao Ono
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.,Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuhiko Nozaki
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Akitsugu Kawashima
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, 477-96 Oowadashinden, Yachiyo City, Chiba, 276-8524, Japan
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1 Kawata-cho, Shinjyuku-ku, Tokyo, 162-8666, Japan
| | - Tomohiro Aoki
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan. .,Core Research for Evolutional Science and Technology from Japan Agency for Medical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita City, Osaka, 564-8565, Japan.
| |
Collapse
|
15
|
Ma S, Zheng J, Xu Y, Yang Z, Zhu Y, Su X, Mo X. Identified plasma proteins related to vascular structure are associated with coarctation of the aorta in children. Ital J Pediatr 2020; 46:63. [PMID: 32430056 PMCID: PMC7236479 DOI: 10.1186/s13052-020-00830-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/11/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coarctation of the aorta (CoA), presenting with local stenosis of the aorta is involved in many cardiovascular processes. However, there has been little research on the mechanism of coarctation of the aorta. METHODS Altered proteins were identified by isobaric tag for relative and absolute quantitation (iTRAQ) technology in 8 participants, and further analysed by heatmap, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) and Search Tool for the Retrieval of Interacting Gene (STRING). Of these, two vascular structure-related proteins were further validated by using enzyme-linked immunosorbent assay (ELISA) in a new cohort of CoA patients. RESULTS 39 differentially expressed plasma proteins were first identified in patients with coarctation of the aorta by iTRAQ. Of these, fibulin-1 (FBLN1) and insulin-like growth factor-binding protein complex acid labile subunit (ALS) were considered candidates and further validation also showed that the level of FBLN1 in the CoA group (8.92 ± 2.36 μg/ml) was significantly higher compared with control group (6.13 ± 1.94 μg/ml), and the level of ALS in CoA children (348.08 ± 216.74 ng/ml) was significantly lower than the level in normal children (619.46 ± 274.08 ng/ml). CONCLUSIONS The differentially expressed proteins identified in the plasma from CoA patients indicated that they may play critical roles in CoA and that they could potentially be utilized as biomarkers for diagnosis. Altered vascular related proteins were associated with COA. These results provide a foundation for further understanding and studying the aetiology and pathogenesis of coarctation of the aorta.
Collapse
Affiliation(s)
- Siyu Ma
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Junqiang Zheng
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yang Xu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yu Zhu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xiaoqi Su
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
16
|
Saraswati S, Lietman CD, Li B, Mathew S, Zent R, Young PP. Small proline-rich repeat 3 is a novel coordinator of PDGFRβ and integrin β1 crosstalk to augment proliferation and matrix synthesis by cardiac fibroblasts. FASEB J 2020; 34:7885-7904. [PMID: 32297675 PMCID: PMC7302973 DOI: 10.1096/fj.201902815r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022]
Abstract
Nearly 6 million Americans suffer from heart failure. Increased fibrosis contributes to functional decline of the heart that leads to heart failure. Previously, we identified a mechanosensitive protein, small proline‐rich repeat 3 (SPRR3), in vascular smooth muscle cells of atheromas. In this study, we demonstrate SPRR3 expression in cardiac fibroblasts which is induced in activated fibroblasts following pressure‐induced heart failure. Sprr3 deletion in mice showed preserved cardiac function and reduced interstitial fibrosis in vivo and reduced fibroblast proliferation and collagen expression in vitro. SPRR3 loss resulted in reduced activation of Akt, FAK, ERK, and p38 signaling pathways, which are coordinately regulated by integrins and growth factors. SPRR3 deletion did not impede integrin‐associated functions including cell adhesion, migration, or contraction. SPRR3 loss resulted in reduced activation of PDGFRβ in fibroblasts. This was not due to the reduced PDGFRβ expression levels or decreased binding of the PDGF ligand to PDGFRβ. SPRR3 facilitated the association of integrin β1 with PDGFRβ and subsequently fibroblast proliferation, suggesting a role in PDGFRβ‐Integrin synergy. We postulate that SPRR3 may function as a conduit for the coordinated activation of PDGFRβ by integrin β1, leading to augmentation of fibroblast proliferation and matrix synthesis downstream of biomechanical and growth factor signals.
Collapse
Affiliation(s)
- Sarika Saraswati
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caressa D Lietman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bin Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sijo Mathew
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, ND, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pampee P Young
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,American Red Cross, Biomedical Division, Washington, DC, USA
| |
Collapse
|
17
|
Maleknia M, Ansari N, Haybar H, Maniati M, Saki N. Inflammatory Growth Factors and In-Stent Restenosis: Effect of Cytokines and Growth Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00240-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
Yılmaz Y, Tanrıverdi F, Duran M, Altay M, Eryol NK. Is IGF-1 level actually lowered in the early stage following an acute myocardial infarction and is IGF-1 associated with the left ventricle dysfunction or cardiac events? JOURNAL OF HEALTH SCIENCES AND MEDICINE 2019. [DOI: 10.32322/jhsm.504184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
19
|
Homozygous receptors for insulin and not IGF-1 accelerate intimal hyperplasia in insulin resistance and diabetes. Nat Commun 2019; 10:4427. [PMID: 31562314 PMCID: PMC6765023 DOI: 10.1038/s41467-019-12368-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/03/2019] [Indexed: 11/08/2022] Open
Abstract
Insulin and IGF-1 actions in vascular smooth muscle cells (VSMC) are associated with accelerated arterial intima hyperplasia and restenosis after angioplasty, especially in diabetes. To distinguish their relative roles, we delete insulin receptor (SMIRKO) or IGF-1 receptor (SMIGF1RKO) in VSMC and in mice. Here we report that intima hyperplasia is attenuated in SMIRKO mice, but not in SMIGF1RKO mice. In VSMC, deleting IGF1R increases homodimers of IR, enhances insulin binding, stimulates p-Akt and proliferation, but deleting IR decreases responses to insulin and IGF-1. Studies using chimeras of IR(extracellular domain)/IGF1R(intracellular-domain) or IGF1R(extracellular domain)/IR(intracellular-domain) demonstrate homodimer IRα enhances insulin binding and signaling which is inhibited by IGF1Rα. RNA-seq identifies hyaluronan synthase2 as a target of homo-IR, with its expression increases by IR activation in SMIGF1RKO mice and decreases in SMIRKO mice. Enhanced intima hyperplasia in diabetes is mainly due to insulin signaling via homo-IR, associated with increased Has2 expression.
Collapse
|
20
|
Guo J, Cui L, Lu Q, Zhang Y, Liu Q, Wang X, Wang Y, Liu Z, Yuan Z, Dai M. Cyadox regulates the transcription of different genes by activation of the PI3K signaling pathway in porcine primary hepatocytes. J Cell Biochem 2019; 120:7623-7634. [PMID: 30417433 DOI: 10.1002/jcb.28037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Cyadox, a new derivative of quinoxalines, has been ascertained as an antibiotic with significant growth promoting, low poison, quick absorption, swift elimination, brief residual period, and noncumulative effect. Seven differential expressed genes, including Insulin-like Growth Factor-1 ( IGF-1), Epidermal Growth Factor ( EGF), Poly ADP-ribose polymerase ( PARP), the Defender Against Apoptotic Death 1 ( DAD1), Complement Component 3 ( C3), Transketolase ( TK) and a New gene, were induced by cyadox in swine liver tissues by messenger RNA differential display reverse transcription polymerase chain reaction (DDRT-PCR) in our laboratory. However, the signal mechanism that cyadox altered these genes expression is not completely elucidated. The signaling pathways involved in the expressions of seven genes induced by cyadox were determined in porcine primary hepatocytes by RT-qPCR and the application of various signal pathway inhibitors. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay revealed that cyadox could stimulate proliferation of porcine primary hepatocytes in a time-dependent manner. In porcine primary cultured hepatocytes, phosphoinositide 3-kinase (PI3K) and transforming growth factor-β (TGF-β) signal pathways were the main signal pathways involved in the expressions of seven genes induced by cyadox. Taken together, these results demonstrate for the first time that seven cyadox-related genes expressions in porcine primary hepatocytes treated with cyadox are mediated mainly through the PI3K signaling pathway, potentially leading to enhanced cell growth and cell immunity. EGF might be the early response gene of cyadox, and a primary regulator of the other gene expressions such as IGF-1 and DAD1, playing an important role in cell proliferation promoted by cyadox.
Collapse
Affiliation(s)
- Ju Guo
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Luqing Cui
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Qirong Lu
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Yinfeng Zhang
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Qianying Liu
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Xu Wang
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Yulian Wang
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Zhenli Liu
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Zonghui Yuan
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| | - Menghong Dai
- MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
21
|
Hooks JS, Clement CC, Nguyen HD, Santambrogio L, Dixon JB. In vitro model reveals a role for mechanical stretch in the remodeling response of lymphatic muscle cells. Microcirculation 2019; 26:e12512. [PMID: 30383330 PMCID: PMC6335159 DOI: 10.1111/micc.12512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/12/2018] [Accepted: 10/29/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Using primary LMCs in vitro, we sought to characterize the impact of LMC remodeling on their functional and molecular response to mechanical loading and culture conditions. METHODS Primary "wounded leg" LMCs were derived from the hindlimb of three sheep who underwent lymphatic injury 6 weeks prior, while "control leg" LMCs were derived from the contralateral, unwounded, limb. Function of the LMCs was characterized in response to media of variable levels of serum (10% vs 0.2%) and glucose (4.5 vs 1 g/L). Functional and proteomic data were evaluated in LMCs exposed to cyclic stretch (0.1 Hz, 7.5% elongation) for 1 week. RESULTS LMCs were sensitive to changes in serum levels, significantly reducing overall activity and collagen synthesis under low serum conditions. LMCs from the remodeled vessel had higher baseline levels of metabolic activity but not collagen synthesis. Cyclic loading induced cellular alignment perpendicular to the axis of stretch and alterations in signaling pathways associated with metabolism. Remodeled LMCs had consistently higher levels of metabolic activity and were more resistant to strain-induced apoptosis. CONCLUSIONS LMCs exist on a functional spectrum, becoming more active in response to stretching and maintaining phenotypic remodeling in response to local lymphatic/tissue damage.
Collapse
Affiliation(s)
- Joshua S.T. Hooks
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology 315 Ferst Dr. Atlanta, GA 30332
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA 30313
| | - Cristina C. Clement
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Hoang-Dung Nguyen
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology 315 Ferst Dr. Atlanta, GA 30332
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA 30332
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - J. Brandon Dixon
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology 315 Ferst Dr. Atlanta, GA 30332
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA 30313
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA 30332
| |
Collapse
|
22
|
Roostalu U, Wong JK. Arterial smooth muscle dynamics in development and repair. Dev Biol 2018; 435:109-121. [PMID: 29397877 DOI: 10.1016/j.ydbio.2018.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/08/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Arterial vasculature distributes blood from early embryonic development and provides a nutrient highway to maintain tissue viability. Atherosclerosis, peripheral artery diseases, stroke and aortic aneurysm represent the most frequent causes of death and are all directly related to abnormalities in the function of arteries. Vascular intervention techniques have been established for the treatment of all of these pathologies, yet arterial surgery can itself lead to biological changes in which uncontrolled arterial wall cell proliferation leads to restricted blood flow. In this review we describe the intricate cellular composition of arteries, demonstrating how a variety of distinct cell types in the vascular walls regulate the function of arteries. We provide an overview of the developmental origin of arteries and perivascular cells and focus on cellular dynamics in arterial repair. We summarize the current knowledge of the molecular signaling pathways that regulate vascular smooth muscle differentiation in the embryo and in arterial injury response. Our review aims to highlight the similarities as well as differences between cellular and molecular mechanisms that control arterial development and repair.
Collapse
Affiliation(s)
- Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK.
| | - Jason Kf Wong
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK; Department of Plastic Surgery, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK.
| |
Collapse
|
23
|
Hoeflich A, David R, Hjortebjerg R. Current IGFBP-Related Biomarker Research in Cardiovascular Disease-We Need More Structural and Functional Information in Clinical Studies. Front Endocrinol (Lausanne) 2018; 9:388. [PMID: 30061864 PMCID: PMC6054974 DOI: 10.3389/fendo.2018.00388] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/25/2018] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death around the world and the insulin-like growth factor (IGF)-system has multiple functions for the pathological conditions of atherosclerosis. IGF binding proteins (IGFBPs) are widely investigated as biomarkers for pathological disorders, including those of the heart. At the tissue level, IGFBP-1 to -6 decrease bioactivity of IGF-I and -II due to their high affinity IGF-binding sites. By contrast, in the circulation, the IGFBPs increase biological half-life of the IGFs and may therefore be regarded as positive regulators of IGF-effects. The IGFBPs may also exert IGF-independent functions inside or outside the cell. Importantly, the circulating IGFBP-concentrations are regulated by trophic, metabolic, and reproductive hormones. In a multitude of studies of healthy subjects and patients with coronary heart diseases, various significant associations between circulating IGFBP-levels and defined parameters have been reported. However, the complex hormonal and conditional control of IGFBPs may explain the lack of clear associations between IGFBPs and parameters of cardiac failure in broader studies including larger populations. Furthermore, the IGFBPs are subject to posttranslational modifications and proteolytic degradation by proteases, upon which the IGFs are released. In this review, we emphasize that, with the exception of IGFBP-4 and in sharp contrast to the preclinical studies, virtually all clinical studies do not have structural or functional information on their biomarker. The use of analytical systems with no discriminatory potential toward intact vs. fragmented IGFBPs represents a major issue in IGFBP-related biomarker research and an important focus point for the future. Overall, measurements of selected IGFBPs or more complex IGFBP-signatures of the family of IGFBPs have potential to identify pathophysiological alterations in the heart or patients with high cardiovascular risk, particularly if defined cohorts are to be assessed. However, a more thorough understanding of the dynamic IGF-IGFBP system as well as its proteases and protease inhibitors in both normal physiology and in cardiovascular diseases is necessary.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Department of Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
- Andreas Hoeflich
| | - Robert David
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany
- Department Life, Light and Matter, Interdisciplinary Faculty, Rostock University, Rostock, Germany
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- The Danish Diabetes Academy, Odense, Denmark
- *Correspondence: Rikke Hjortebjerg
| |
Collapse
|
24
|
Masiero G, Mojoli M, Ueshima D, Tarantini G. Current concepts on coronary revascularization using BRS in patients with diabetes and small vessels disease. J Thorac Dis 2017; 9:S940-S949. [PMID: 28894600 PMCID: PMC5583088 DOI: 10.21037/jtd.2017.06.36] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/17/2017] [Indexed: 01/19/2023]
Abstract
Diabetes mellitus (DM) and small vessel (SV) disease are two major predictors of adverse outcome in patients treated by percutaneous coronary intervention (PCI), even when last generation metallic drug-eluting stents (DES) are used. Bioresorbable scaffold (BRS) technology has been recently developed to overcome the disadvantages of metallic DES due to their permanent struts. Through the resorption process, BRS may provide a vascular restoration that appears very attractive especially when distal or diffusely diseased coronary segments are involved, as in diabetic patients and SV disease. However, robust evidence on the use of BRS in diabetics is lacking, and recent data have raised concerns on the use of BRS in SVs, particularly when reference vessel diameter (RVD) is <2.25 mm. This review aims at summarizing current evidence related to the use of BRS in diabetics and SV disease.
Collapse
Affiliation(s)
- Giulia Masiero
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padua, Italy
| | - Marco Mojoli
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padua, Italy
| | - Daisuke Ueshima
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padua, Italy
| | - Giuseppe Tarantini
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padua, Italy
| |
Collapse
|
25
|
Beneit N, Fernández-García CE, Martín-Ventura JL, Perdomo L, Escribano Ó, Michel JB, García-Gómez G, Fernández S, Díaz-Castroverde S, Egido J, Gómez-Hernández A, Benito M. Expression of insulin receptor (IR) A and B isoforms, IGF-IR, and IR/IGF-IR hybrid receptors in vascular smooth muscle cells and their role in cell migration in atherosclerosis. Cardiovasc Diabetol 2016; 15:161. [PMID: 27905925 PMCID: PMC5134076 DOI: 10.1186/s12933-016-0477-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 01/02/2023] Open
Abstract
Background Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is a major contributor to the development of atherosclerotic process. In a previous work, we demonstrated that the insulin receptor isoform A (IRA) and its association with the insulin-like growth factor-I receptor (IGF-IR) confer a proliferative advantage to VSMCs. However, the role of IR and IGF-IR in VSMC migration remains poorly understood. Methods Wound healing assays were performed in VSMCs bearing IR (IRLoxP+/+ VSMCs), or not (IR−/− VSMCs), expressing IRA (IRA VSMCs) or expressing IRB (IRB VSMCs). To study the role of IR isoforms and IGF-IR in experimental atherosclerosis, we used ApoE−/− mice at 8, 12, 18 and 24 weeks of age. Finally, we analyzed the mRNA expression of total IR, IRB isoform, IGF-IR and IGFs by qRT-PCR in the medial layer of human aortas. Results IGF-I strongly induced migration of the four cell lines through IGF-IR. In contrast, insulin and IGF-II only caused a significant increase of IRA VSMC migration which might be favored by the formation of IRA/IGF-IR receptors. Additionally, a specific IGF-IR inhibitor, picropodophyllin, completely abolished insulin- and IGF-II-induced migration in IRB, but not in IRA VSMCs. A significant increase of IRA and IGF-IR, and VSMC migration were observed in fibrous plaques from 24-week-old ApoE−/− mice. Finally, we observed a marked increase of IGF-IR, IGF-I and IGF-II in media from fatty streaks as compared with both healthy aortas and fibrolipidic lesions, favoring the ability of medial VSMCs to migrate into the intima. Conclusions Our data suggest that overexpression of IGF-IR or IRA isoform, as homodimers or as part of IRA/IGF-IR hybrid receptors, confers a stronger migratory capability to VSMCs as might occur in early stages of atherosclerotic process. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0477-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- N Beneit
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - C E Fernández-García
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - J L Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - L Perdomo
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Ó Escribano
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J B Michel
- Inserm, U698, Universite Paris 7, CHU X-Bichat, Paris, France
| | - G García-Gómez
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Fernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Díaz-Castroverde
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J Egido
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.,Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - A Gómez-Hernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain. .,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain. .,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.
| | - M Benito
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| |
Collapse
|
26
|
Abstract
Type 2 diabetes mellitus is increasing in prevalence, and is a potent risk factor for the development of atherosclerotic vascular disease and increased risk of adverse cardiovascular events. Approximately 15—25% of patients presenting with ischaemic heart disease have a history of diabetes mellitus. This cohort of patients continues to be at heightened short and long-term risk. This review highlights the many proposed biological drivers that likely play a deterministic role in the cardiovascular outcome of patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Steven P Marso
- The Mid America Heart Institute, Saint Lukes Hospital, 4401 Wornall, Kansas City, Missouri 64111, USA,
| |
Collapse
|
27
|
Zhao J, Schnitzler GR, Iyer LK, Aronovitz MJ, Baur WE, Karas RH. MicroRNA-Offset RNA Alters Gene Expression and Cell Proliferation. PLoS One 2016; 11:e0156772. [PMID: 27276022 PMCID: PMC4898817 DOI: 10.1371/journal.pone.0156772] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/19/2016] [Indexed: 11/29/2022] Open
Abstract
MicroRNA-offset RNAs (moRs) were first identified in simple chordates and subsequently in mouse and human cells by deep sequencing of short RNAs. MoRs are derived from sequences located immediately adjacent to microRNAs (miRs) in the primary miR (pri-miR). Currently moRs are considered to be simply a by-product of miR biosynthesis that lack biological activity. Here we show for the first time that a moR is biologically active. We demonstrate that endogenous or over-expressed moR-21 significantly alters gene expression and inhibits the proliferation of vascular smooth muscle cells (VSMC). In addition, we find that miR-21 and moR-21 may regulate different genes in a given pathway and can oppose each other in regulating certain genes. We report that there is a “seed region” of moR-21 as well as a “seed match region” in the target gene 3’UTR that are indispensable for moR-21-mediated gene down-regulation. We further demonstrate that moR-21-mediated gene repression is Argonaute 2 (Ago2) dependent. Taken together, these findings provide the first evidence that microRNA offset RNA alters gene expression and is biologically active.
Collapse
Affiliation(s)
- Jin Zhao
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
| | - Gavin R. Schnitzler
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
| | - Lakshmanan K. Iyer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
| | - Mark J. Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
| | - Wendy E. Baur
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
| | - Richard H. Karas
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, United States of America
- * E-mail:
| |
Collapse
|
28
|
Magnobovatol inhibits smooth muscle cell migration by suppressing PDGF-Rβ phosphorylation and inhibiting matrix metalloproteinase-2 expression. Int J Mol Med 2016; 37:1239-46. [PMID: 27049716 PMCID: PMC4829143 DOI: 10.3892/ijmm.2016.2548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/29/2016] [Indexed: 11/23/2022] Open
Abstract
The migration of vascular smooth muscle cells (VSMCs) may play a crucial role in the pathogenesis of vascular diseases, such as atherosclerosis and post-angioplasty restenosis. Platelet-derived growth factor (PDGF)-BB is a potent mitogen for VSMCs and plays an important role in the intimal accumulation of VSMCs. Magnobovatol, a new neolignan from the fruits of Magnolia obovata, has been shown to have anticancer properties. However, the effects of magnobovatol on VSMCs are unknown. In the present study, we examined the effects of magnobovatol on the PDGF-BB-induced migration of mouse and human VSMCs, as well as the underlying mechanisms. Magnobovatol significantly inhibited the PDGF-BB-induced migration of mouse and human VSMCs without inducing cell death (as shown by MTT assay and wound healing assay). Additionally, we demonstrated that magnobovatol significantly blocked the PDGF-BB-induced phosphorylation of the PDGF receptor (PDGF-R), Akt and extracellular signal-regulated kinase (ERK)1/2 by inhibiting the activation of the PDGF-BB signaling pathway. Moreover, in both mouse and human VSMCs, magnobovatol inhibited PDGF-induced matrix metalloproteinase (MMP)-2 expression at the mRNA and protein level, as well as the proteolytic activity of MMP-2 (as shown by western blot analysis, RT-PCR, gelatin zymography and ELISA). In addition, the sprout outgrowth formation of aortic rings induced by PDGF-BB was inhibited by magnobovatol (as shown by aortic ring assay). Taken together, our findings indicate that magnobovatol inhibits VSMC migration by decreasing MMP-2 expression through PDGF-R and the ERK1/2 and Akt pathways. Our data may improve the understanding of the anti-atherogenic effects of magnobovatol in VSMCs.
Collapse
|
29
|
Bai X, Huang L, Hu K, Qu F. Inhibited proliferation of human umbilical artery smooth muscle cells by xanthinol nicotinate. Med Biol Eng Comput 2016; 54:891-8. [PMID: 26718554 DOI: 10.1007/s11517-015-1438-9] [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: 11/30/2013] [Accepted: 12/12/2015] [Indexed: 11/26/2022]
Abstract
Vascular smooth muscle cell proliferation is a key event in the development of hypertension, instant restenosis and other cardiac disorders. Inhibition of this proliferation could lead to better prevention and treatment of these diseases. This study was designed to investigate the effects and mechanisms of different concentrations of xanthinol nicotinate (XN) on human umbilical artery smooth muscle cell (HUASMC) proliferation in vitro. HUASMCs were cultured by the tissue adherent method, passaged three times, and then identified by immunohistochemistry. HUASMCs were then treated with different concentrations of XN (0, 2.76, 27.6 or 276 µM), and a 3-(4,5-dimethylthiazol-2yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was used to detect the inhibition of HUASMC proliferation. The levels of platelet-derived growth factor receptor (PDGFR) mRNA and protein (PDGFR-β) were detected on the cell membrane of these treated HUASMCs using RT-PCR and Western blot analysis, respectively. After culturing and passaging three times, 90 % of the cultured cells were identified as HUASMCs by immunohistochemistry. HUASMC proliferation was inhibited by XN in a dose-dependent manner (P < 0.05). Furthermore, XN dose-dependently decreased the PDGFR mRNA and PDGFR-β levels on the cell membranes of HUASMCs (P < 0.05). Thus, the results suggest that XN could become a potent therapeutic agent for regulating VSMC-associated vascular disease such as cardiovascular disease and restenosis after angioplasty.
Collapse
Affiliation(s)
- Xiaodan Bai
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, Heilongjiang Province, China
- Department of Pharmacy, Harbin Traditional Chinese Medical Hospital, 270 Jianguo Street, Daoli District, Harbin, 150076, Heilongjiang Province, China
| | - Lijun Huang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Kejie Hu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, Heilongjiang Province, China
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Fujun Qu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, Heilongjiang Province, China.
| |
Collapse
|
30
|
Awasthi N, Schwarz RE. Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco Targets Ther 2015; 8:3691-701. [PMID: 26677336 PMCID: PMC4677757 DOI: 10.2147/ott.s78805] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is an essential process for tumor growth and metastasis, and remains a promising therapeutic target process in cancer treatment for several cancer types. Bevacizumab, a monoclonal antibody that targets vascular endothelial growth factor (VEGF), was the first antiangiogenic agent approved for cancer therapy. Novel antiangiogenic agents, such as sunitinib, sorafenib, pazopanib, or vandetanib that target additional proangiogenic signaling pathways beyond VEGF, have also been approved for the treatment of various malignant diseases. While most of these agents are approved in combination with cytotoxic chemotherapy for indications including metastatic colorectal cancer, non-small-cell lung cancer, breast cancer, renal cell carcinoma (RCC), and gastric cancer, some are used as approved monotherapy for advanced RCC, hepatocellular carcinoma and medullary thyroid cancer. Major challenges to the success of antiangiogenic therapy include associated toxicity risks, limitation of efficacy through the possible development of resistance and induction or promotion of metastatic progression. Nintedanib (formally known as BIBF 1120) is a triple angiokinase inhibitor of VEGF, fibroblast growth factor, platelet-derived growth factor signaling with lesser activity against RET, Flt-3, and Src. Through this unique targeting profile nintedanib has demonstrated significant antitumor activity in several tumor types in preclinical studies. Nintedanib has also shown promising clinical efficacy in combination with docetaxel and has been approved for treating patients with locally advanced and metastatic non-small-cell lung cancer in Europe. Nintedanib has also been found to be clinically promising in terms of efficacy and safety in several other solid tumors including ovarian cancer (Phase III), RCC (Phase II), and prostate cancer (Phase II). This review article provides a comprehensive summary of the preclinical and clinical efficacy of nintedanib in the treatment of solid tumors.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA ; Indiana University Health Goshen Center for Cancer Care, Goshen, IN, USA
| |
Collapse
|
31
|
Gomez D, Swiatlowska P, Owens GK. Epigenetic Control of Smooth Muscle Cell Identity and Lineage Memory. Arterioscler Thromb Vasc Biol 2015; 35:2508-16. [PMID: 26449751 PMCID: PMC4662608 DOI: 10.1161/atvbaha.115.305044] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/23/2015] [Indexed: 12/31/2022]
Abstract
Vascular smooth muscle cells (SMCs), like all cells, acquire a cell-specific epigenetic signature during development that includes acquisition of a unique repertoire of histone and DNA modifications. These changes are postulated to induce an open chromatin state (referred to as euchromatin) on the repertoire of genes that are expressed in differentiated SMC, including SMC-selective marker genes like Acta2 and Myh11, as well as housekeeping genes expressed by most cell types. In contrast, genes that are silenced in differentiated SMC acquire modifications associated with a closed chromatin state (ie, heterochromatin) and transcriptional silencing. Herein, we review mechanisms that regulate epigenetic control of the differentiated state of SMC. In addition, we identify some of the major limitations in the field and future challenges, including development of innovative new tools and approaches, for performing single-cell epigenetic assays and locus-selective editing of the epigenome that will allow direct studies of the functional role of specific epigenetic controls during development, injury repair, and disease, including major cardiovascular diseases, such as atherosclerosis, hypertension, and microvascular disease, associated with diabetes mellitus.
Collapse
MESH Headings
- Animals
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cardiovascular Diseases/physiopathology
- Cell Differentiation/genetics
- Cell Lineage/drug effects
- Chromatin Assembly and Disassembly
- Embryonic Stem Cells/metabolism
- Embryonic Stem Cells/pathology
- Epigenesis, Genetic
- Epigenomics/methods
- Gene Expression Regulation, Developmental
- Genetic Markers
- Humans
- Muscle Development/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
Collapse
Affiliation(s)
- Delphine Gomez
- From the Department of Molecular Physiology and Biological Physics (D.G., G.K.O.), and Robert M. Berne Cardiovascular Research Center (P.S.), University of Virginia School of Medicine, Charlottesville
| | - Pamela Swiatlowska
- From the Department of Molecular Physiology and Biological Physics (D.G., G.K.O.), and Robert M. Berne Cardiovascular Research Center (P.S.), University of Virginia School of Medicine, Charlottesville
| | - Gary K Owens
- From the Department of Molecular Physiology and Biological Physics (D.G., G.K.O.), and Robert M. Berne Cardiovascular Research Center (P.S.), University of Virginia School of Medicine, Charlottesville.
| |
Collapse
|
32
|
Molecular Pathways Regulating Macrovascular Pathology and Vascular Smooth Muscle Cells Phenotype in Type 2 Diabetes. Int J Mol Sci 2015; 16:24353-68. [PMID: 26473856 PMCID: PMC4632754 DOI: 10.3390/ijms161024353] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/21/2015] [Accepted: 10/08/2015] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a disease reaching a pandemic proportion in developed countries and a major risk factor for almost all cardiovascular diseases and their adverse clinical manifestations. T2DM leads to several macrovascular and microvascular alterations that influence the progression of cardiovascular diseases. Vascular smooth muscle cells (VSMCs) are fundamental players in macrovascular alterations of T2DM patients. VSMCs display phenotypic and functional alterations that reflect an altered intracellular biomolecular scenario of great vessels of T2DM patients. Hyperglycemia itself and through intraparietal accumulation of advanced glycation-end products (AGEs) activate different pathways, in particular nuclear factor-κB and MAPKs, while insulin and insulin growth-factor receptors (IGFR) are implicated in the activation of Akt and extracellular-signal-regulated kinases (ERK) 1/2. Nuclear factor-κB is also responsible of increased susceptibility of VSMCs to pro-apoptotic stimuli. Down-regulation of insulin growth-factor 1 receptors (IGFR-1R) activity in diabetic vessels also influences negatively miR-133a levels, so increasing apoptotic susceptibility of VSMCs. Alterations of those bimolecular pathways and related genes associate to the prevalence of a synthetic phenotype of VSMCs induces extracellular matrix alterations of great vessels. A better knowledge of those biomolecular pathways and related genes in VSMCs will help to understand the mechanisms leading to macrovascular alterations in T2DM patients and to suggest new targeted therapies.
Collapse
|
33
|
Kubohara Y, Komachi M, Homma Y, Kikuchi H, Oshima Y. Derivatives of Dictyostelium differentiation-inducing factors inhibit lysophosphatidic acid-stimulated migration of murine osteosarcoma LM8 cells. Biochem Biophys Res Commun 2015; 463:800-5. [PMID: 26056940 DOI: 10.1016/j.bbrc.2015.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 10/23/2022]
Abstract
Osteosarcoma is a common metastatic bone cancer that predominantly develops in children and adolescents. Metastatic osteosarcoma remains associated with a poor prognosis; therefore, more effective anti-metastatic drugs are needed. Differentiation-inducing factor-1 (DIF-1), -2, and -3 are novel lead anti-tumor agents that were originally isolated from the cellular slime mold Dictyostelium discoideum. Here we investigated the effects of a panel of DIF derivatives on lysophosphatidic acid (LPA)-induced migration of mouse osteosarcoma LM8 cells by using a Boyden chamber assay. Some DIF derivatives such as Br-DIF-1, DIF-3(+2), and Bu-DIF-3 (5-20 μM) dose-dependently suppressed LPA-induced cell migration with associated IC50 values of 5.5, 4.6, and 4.2 μM, respectively. On the other hand, the IC50 values of Br-DIF-1, DIF-3(+2), and Bu-DIF-3 versus cell proliferation were 18.5, 7.2, and 2.0 μM, respectively, in LM8 cells, and >20, 14.8, and 4.3 μM, respectively, in mouse 3T3-L1 fibroblasts (non-transformed). Together, our results demonstrate that Br-DIF-1 in particular may be a valuable tool for the analysis of cancer cell migration, and that DIF derivatives such as DIF-3(+2) and Bu-DIF-3 are promising lead anti-tumor agents for the development of therapies that suppress osteosarcoma cell proliferation, migration, and metastasis.
Collapse
Affiliation(s)
- Yuzuru Kubohara
- Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan; Department of Health Science, Juntendo University Graduate School of Health and Sports Science, Inzai 270-1695, Japan.
| | - Mayumi Komachi
- Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan; Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yoshimi Homma
- Department of Biomolecular Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Haruhisa Kikuchi
- Laboratory of Natural Product Chemistry, Tohoku University Graduate School of Pharmaceutical Sciences, Aoba-yama, Aoba-ku, Sendai 980-8578, Japan
| | - Yoshiteru Oshima
- Laboratory of Natural Product Chemistry, Tohoku University Graduate School of Pharmaceutical Sciences, Aoba-yama, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
34
|
Kim DY, Won KJ, Yoon MS, Yu HJ, Park JH, Kim B, Lee HM. Chrysanthemum boreale flower floral water inhibits platelet-derived growth factor-stimulated migration and proliferation in vascular smooth muscle cells. PHARMACEUTICAL BIOLOGY 2015; 53:725-734. [PMID: 25330930 DOI: 10.3109/13880209.2014.941882] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Chrysanthemum boreale Makino (Compositae) (CBM) is a traditional medicine that has been used for the prevention or treatment of various disorders; it has various properties including antioxidation, anti-inflammation, and antitumor. OBJECTIVE The present study was designed to explore the in vitro effect of CBM flower floral water (CBMFF) on atherosclerosis-related responses in rat aortic smooth muscle cells (RASMCs). MATERIALS AND METHODS CBMFF was extracted from CBM flower by steam distillation and analyzed using gas chromatography-mass spectrometry. The anti-atherosclerosis activity of CBMFF was tested by estimating platelet-derived growth factor (PDGF)-BB (10 ng/mL)-induced proliferation and migration levels and intracellular kinase pathways in RASMCs at CBMFF concentrations of 0.01-100 μM and analyzing ex vivo aortic ring assay. RESULTS Gas chromatography-mass spectrometry showed that the CBMFF contained a total of seven components. The CBMFF inhibits PDGF-BB-stimulated RASMC migration and proliferation (IC50: 0.010 μg/mL). Treatment of RASMCs with PDGF-BB induced PDGFR-β phosphorylation and increased the phosphorylations of MAPK p38 and ERK1/2. CBMFF addition prevented PDGF-BB-induced phosphorylation of these kinases (IC50: 008 and 0.018 μg/mL, for p38 MAPK and ERK1/2, respectively), as well as PDGFR-β (IC50: 0.046 μg/mL). Treatment with inhibitors of PDGFR, P38 MAPK, and ERK1/2 decreased PDGF-BB-increased migration and proliferation in RASMCs. Moreover, the CBMFF suppressed PDGF-BB-increased sprout outgrowth of aortic rings (IC50: 0.047 μg/mL). DISCUSSION AND CONCLUSION These results demonstrate that CBMFF may inhibit PDGF-BB-induced vascular migration and proliferation, most likely through inhibition of the PDGFR-β-mediated MAPK pathway; therefore, the CBMFF may be promising candidate for the development of herbal remedies for vascular disorders.
Collapse
MESH Headings
- Animals
- Cell Movement/drug effects
- Cell Movement/physiology
- Cell Proliferation/drug effects
- Cell Proliferation/physiology
- Cells, Cultured
- Chrysanthemum
- Dose-Response Relationship, Drug
- Flowers
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/pharmacology
- Rats
- Rats, Sprague-Dawley
- Water/pharmacology
Collapse
Affiliation(s)
- Do-Yoon Kim
- Department of Cosmetic Science, College of Natural Science, Hoseo University , Asan, Chungnam Prefecture , Republic of Korea and
| | | | | | | | | | | | | |
Collapse
|
35
|
Mitchell A, Fujisawa T, Newby D, Mills N, Cruden NL. Vascular injury and repair: a potential target for cell therapies. Future Cardiol 2015; 11:45-60. [DOI: 10.2217/fca.14.77] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
ABSTRACT Whether due to atherosclerotic disease or mechanical intervention, vascular injury is a frequently encountered pathology in cardiovascular medicine. The past decade has seen growing interest in the role of circulating endothelial progenitor cells in vessel recovery postinjury. Despite this, the definition, origin and potential role of endothelial progenitor cells in vascular regeneration remains highly controversial. While animal work has shown early promise, evidence of a therapeutic role for endothelial progenitor cells in humans remains elusive. To date, clinical trials involving direct cell administration, growth factor therapy and endothelial cell capture stents have largely been disappointing, although this may in part reflect limitations in study design. This article will outline the pathophysiological mechanisms of vascular injury with an emphasis on endothelial progenitor cell biology and the potential therapeutic role of this exciting new field.
Collapse
Affiliation(s)
- Andrew Mitchell
- Centre for Cardiovascular Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Takeshi Fujisawa
- Scottish Centre for Regenerative Medicine; Edinburgh Bioquarter; 5 Little France Drive, Edinburgh, UK
| | - David Newby
- Centre for Cardiovascular Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Nicholas Mills
- Centre for Cardiovascular Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Nicholas L Cruden
- Centre for Cardiovascular Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| |
Collapse
|
36
|
Lee KP, Park ES, Kim DE, Park IS, Kim JT, Hong H. Artemisinin attenuates platelet-derived growth factor BB-induced migration of vascular smooth muscle cells. Nutr Res Pract 2014; 8:521-5. [PMID: 25324931 PMCID: PMC4198964 DOI: 10.4162/nrp.2014.8.5.521] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/14/2014] [Accepted: 07/25/2014] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND/OBJECTIVES Artemisinin (AT), an active compound in Arternisia annua, is well known as an anti-malaria drug. It is also known to have several effects including anti-oxidant, anti-inflammation, and anti-cancer activities. To date, the effect of AT on vascular disorders has not been studied. In this study, we investigated the effects of AT on the migration and proliferation of vascular smooth muscle cells (VSMC) stimulated by platelet-derived growth factor BB (PDGF-BB). MATERIALS/METHODS Aortic smooth muscle cells were isolated from Sprague-Dawley rats. PDGF-BB stimulated VSMC migration was measured by the scratch wound healing assay and the Boyden chamber assay. Cell viability was determined by using an EZ-Cytox Cell Viability Assay Kit. The production of reactive oxygen species (ROS) in PDGF-BB stimulated VSMC was measured through H2DCF-DA staining. We also determined the expression levels of signal proteins relevant to ROS, including measures of extracellular signal-regulated kinase (ERK) 1/2 measured by western blot analysis and matrix metalloproteinase (MMP) 9 measured by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS AT (10 µM and 30 µM) significantly reduced the proliferation and migration of PDGF-BB stimulated VSMC in a dose-dependent manner. The production of ROS, normally induced by PDGF-BB, is reduced by treatment with AT at both concentrations. PDGF-BB stimulated VSMC treated with AT (10 µM and 30 µM) have reduced phosphorylation of ERK1/2 and inhibited MMP9 expression compared to untreated PDGF-BB stimulated VSMC. CONCLUSIONS We suggest, based on these results, that AT may exert an anti-atherosclerotic effect on PDGF-BB stimulated VSMCs by inhibiting their proliferation and migration through down-regulation of ERK1/2 and MMP9 phosphorylation.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Medical Science, School of Medicine Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea
| | - Eun-Seok Park
- Department of Biomedical Laboratory Science, Kyungbok University, Gyeonggi-do 487-717, Korea
| | - Dae-Eun Kim
- Department of Biomedical Laboratory Science, Kyungbok University, Gyeonggi-do 487-717, Korea
| | - In-Sik Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongsangbuk-do 780-714, Korea
| | - Jin Tack Kim
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongsangbuk-do 780-714, Korea
| | - Heeok Hong
- Department of Medical Science, School of Medicine Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea
| |
Collapse
|
37
|
Iñiguez G, Castro JJ, Garcia M, Kakarieka E, Johnson MC, Cassorla F, Mericq V. IGF-IR signal transduction protein content and its activation by IGF-I in human placentas: relationship with gestational age and birth weight. PLoS One 2014; 9:e102252. [PMID: 25050889 PMCID: PMC4106823 DOI: 10.1371/journal.pone.0102252] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/16/2014] [Indexed: 02/05/2023] Open
Abstract
Introduction The human placenta expresses the IGF-I and IGF-IR proteins and their intracellular signal components (IRS-1, AKT and mTOR). The aim of this study was to assess the IGF-IR content and activation of downstream signaling molecules in placentas from newborns who were classified by gestational age and birth weight. We studied placentas from 25 term appropriate (T-AGA), 26 term small (T-SGA), 22 preterm AGA (PT-AGA), and 20 preterm SGA (PT-SGA) newborns. The total and phosphorylated IGF-IR, IRS-1, AKT, and mTOR contents were determined by Western Blot and normalized by actin or with their respective total content. The effect of IGF-I was determined by stimulating placental explants with recombinant IGF-I 10-8 mol/L for 15, 30, and 60 minutes. Results The IGF-IR content was higher in T-SGA compared to T-AGA placentas, and the IRS-1 content was higher in PT-placentas compared with their respective T-placentas. The effect of IGF-I on the phosphorylated forms of IGF-IR was increased in T-SGA (150%) and PT-SGA (300%) compared with their respective AGA placentas. In addition, AKT serine phosphorylation was higher in PT-SGA compared to PT-AGA and T-SGA placentas (90% and 390% respectively). Conclusion The higher protein content and response to IGF-I of IGF-IR, IRS-1, and AKT observed in SGA placentas may represent a compensatory mechanism in response to fetal growth restriction.
Collapse
Affiliation(s)
- Germán Iñiguez
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
- * E-mail:
| | - Juan José Castro
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Mirna Garcia
- Hospital Clínico San Borja-Arriarán, University of Chile, Santiago, Chile
| | - Elena Kakarieka
- Hospital Clínico San Borja-Arriarán, University of Chile, Santiago, Chile
| | - M. Cecilia Johnson
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Fernando Cassorla
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Verónica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| |
Collapse
|
38
|
1-year clinical outcomes of diabetic patients treated with everolimus-eluting bioresorbable vascular scaffolds: a pooled analysis of the ABSORB and the SPIRIT trials. JACC Cardiovasc Interv 2014; 7:482-93. [PMID: 24746650 DOI: 10.1016/j.jcin.2014.01.155] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/30/2013] [Accepted: 01/04/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate 1-year clinical outcomes of diabetic patients treated with the Absorb bioresorbable vascular scaffold (BVS). BACKGROUND Clinical outcomes of diabetic patients after BVS implantation have been unreported. METHODS This study included 101 patients in the ABSORB Cohort B trial and the first consecutive 450 patients with 1 year of follow-up in the ABSORB EXTEND trial. A total of 136 diabetic patients were compared with 415 nondiabetic patients. In addition, 882 diabetic patients treated with everolimus-eluting metal stents (EES) in pooled data from the SPIRIT trials (SPIRIT FIRST [Clinical Trial of the Abbott Vascular XIENCE V Everolimus Eluting Coronary Stent System], SPIRIT II [A Clinical Evaluation of the XIENCE V Everolimus Eluting Coronary Stent System], SPIRIT III [Clinical Trial of the XIENCE V Everolimus Eluting Coronary Stent System (EECSS)], SPIRIT IV Clinical Trial [Clinical Evaluation of the XIENCE V Everolimus Eluting Coronary Stent System]) were used for the comparison by applying propensity score matching. The primary endpoint was a device-oriented composite endpoint (DoCE), including cardiac death, target vessel myocardial infarction, and target lesion revascularization at 1-year follow-up. RESULTS The cumulative incidence of DoCE did not differ between diabetic and nondiabetic patients treated with the BVS (3.7% vs. 5.1%, p = 0.64). Diabetic patients treated with the BVS had a similar incidence of the DoCE compared with diabetic patients treated with EES in the matched study group (3.9% for the BVS vs. 6.4% for EES, p = 0.38). There were no differences in the incidence of definite or probable scaffold/stent thrombosis (0.7% for both diabetic and nondiabetic patients with the BVS; 1.0% for diabetic patients with the BVS vs. 1.7% for diabetic patients with EES in the matched study group). CONCLUSIONS In the present analyses, diabetic patients treated with the BVS showed similar rates of DoCEs compared with nondiabetic patients treated with the BVS and diabetic patients treated with EES at 1-year follow-up. (ABSORB Clinical Investigation, Cohort B; NCT00856856; ABSORB EXTEND Clinical Investigation; NCT01023789; Clinical Trial of the Abbott Vascular XIENCE V Everolimus Eluting Coronary Stent System [SPIRIT FIRST]; NCT00180453; A Clinical Evaluation of the XIENCE V Everolimus Eluting Coronary Stent System [SPIRIT II]; NCT00180310; Clinical Trial of the XIENCE V Everolimus Eluting Coronary Stent System [EECSS] [SPIRIT III]; NCT00180479; Clinical Evaluation of the XIENCE V Everolimus Eluting Coronary Stent System [SPIRIT IV Clinical Trial]; NCT00307047).
Collapse
|
39
|
Wagner B, Gorin Y. Src tyrosine kinase mediates platelet-derived growth factor BB-induced and redox-dependent migration in metanephric mesenchymal cells. Am J Physiol Renal Physiol 2013; 306:F85-97. [PMID: 24197068 DOI: 10.1152/ajprenal.00371.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The adult kidney is derived from the interaction between the metanephric blastema and the ureteric bud. Platelet-derived growth factor (PDGF) receptor β is essential for the development of the mature glomerular tuft, as mice deficient for this receptor lack mesangial cells. This study investigated the role of Src tyrosine kinase in PDGF-mediated reactive oxygen species (ROS) generation and migration of metanephric mesenchymal cells (MMCs). Cultured embryonic MMCs from wild-type and PDGF receptor-deficient embryos were established. Migration was determined via wound-healing assay. Unlike PDGF AA, PDGF BB-induced greater migration in MMCs with respect to control. This was abrogated by neutralizing an antibody to PDGF BB. Phosphatidylinositol 3-kinase (PI3K) inhibitors suppressed PDGF BB-induced migration. Conversely, mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors had no effect. Src inhibitors inhibited PDGF-induced cell migration, PI3K activity, and Akt phosphorylation. Adenoviral dominant negative Src (AD DN Src) abrogated PDGF BB-induced Akt phosphorylation. Hydrogen peroxide stimulated cell migration. PDGF BB-induced wound closure was inhibited by the antioxidants N-acetyl-l-cysteine, tiron, and the flavoprotein inhibitor diphenyleneiodonium. These cells express the NADPH oxidase homolog Nox4. Inhibiting Nox4 with antisense oligonucleotides or small interfering RNA (siRNA) suppressed PDGF-induced wound closure. Inhibition of Src with siRNA reduced PDGF BB-induced ROS generation as assessed by 2',7'-dichlorodihydrofluorescein diacetate fluorescence. Furthermore, PDGF BB-stimulated ROS generation and migration were similarly suppressed by Ad DN Src. In MMCs, PDGF BB-induced migration is mediated by PI3K and Src in a redox-dependent manner involving Nox4. Src may be upstream to PI3K and Nox4.
Collapse
Affiliation(s)
- Brent Wagner
- South Texas Veterans Health Care System, Div. of Nephrology MC 7882, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900.
| | | |
Collapse
|
40
|
Chen YF, Tsai HY, Wu KJ, Siao LR, Wood WG. Pipoxolan ameliorates cerebral ischemia via inhibition of neuronal apoptosis and intimal hyperplasia through attenuation of VSMC migration and modulation of matrix metalloproteinase-2/9 and Ras/MEK/ERK signaling pathways. PLoS One 2013; 8:e75654. [PMID: 24086601 PMCID: PMC3782448 DOI: 10.1371/journal.pone.0075654] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/16/2013] [Indexed: 12/05/2022] Open
Abstract
Pipoxolan (PIPO) has anti-spasmodic effects, and it is used clinically to relieve smooth muscle spasms. Cerebrovascular disease is one of the leading causes of disability and death worldwide. The main aim of this study was to investigate the effects of PIPO on cerebral ischemia and vascular smooth muscle cell (VSMC) migration in vivo and in vitro. Cerebral infarction area, ratio of intima to media area (I/M ratio) and PCNA antibody staining of the carotid artery in vivo were measured. Cell viability of A7r5 cells, PDGF-BB-stimulated cell migration, and potential mechanisms of PIPO were evaluated by wound healing, transwell and Western blotting. PIPO (10 and 30 mg/kg p.o.) reduced: the cerebral infarction area; neurological deficit; TUNEL-positive cells; cleaved caspase 3-positive cells; intimal hyperplasia; and inhibited proliferating cell nuclear antigen (PCNA)-positive cells in rodents. PIPO (5, 10 and 15 µM) significantly inhibited PDGF-BB-stimulated VSMC migration and reduced Ras, MEK, and p-ERK levels. Moreover, PIPO decreased levels of matrix metalloproteinases -2 and -9 in PDGF-BB-stimulated A7r5 cells. In summary, PIPO is protective in models of ischemia/reperfusion-induced cerebral infarction, carotid artery ligation-induced intimal hyperplasia and VSMC migration both in vivo and in vitro. PIPO could be potentially efficacious in preventing cerebrovascular and vascular diseases.
Collapse
Affiliation(s)
- Yuh-Fung Chen
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
- * E-mail:
| | - Huei-Yann Tsai
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Kuo-Jen Wu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Lian-Ru Siao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - W. Gibson Wood
- Department of Pharmacology, University of Minnesota and Geriatric Research, Education and Clinical Center, VA Medical Center, Minneapolis, Minnesota, United States of America
| |
Collapse
|
41
|
Turner EC, Huang CL, Govindarajan K, Caplice NM. Identification of a Klf4-dependent upstream repressor region mediating transcriptional regulation of the myocardin gene in human smooth muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1191-201. [PMID: 24060351 DOI: 10.1016/j.bbagrm.2013.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/28/2013] [Accepted: 09/13/2013] [Indexed: 01/25/2023]
Abstract
Phenotypic switching of smooth muscle cells (SMCs) plays a central role in the development of vascular diseases such as atherosclerosis and restenosis. However, the factors regulating expression of the human myocardin (Myocd) gene, the master gene regulator of SMC differentiation, have yet to be identified. In this study, we sought to identify the critical factors regulating Myocd expression in human SMCs. Using deletion/genetic reporter analyses, an upstream repressor region (URR) was localised within the Myocd promoter, herein termed PrmM. Bioinformatic analysis revealed three evolutionary conserved Klf4 sites within the URR and disruption of those elements led to substantial increases in PrmM-directed gene expression. Furthermore, ectopic expression established that Klf4 significantly decreased Myocd mRNA levels and PrmM-directed gene expression while electrophoretic mobility shift assays and chromatin immunoprecipitation (ChIP) assays confirmed specific binding of endogenous Klf4, and not Klf5 or Klf2, to the URR of PrmM. Platelet-derived growth factor BB (PDGF-BB), a potent inhibitor of SMC differentiation, reduced Myocd mRNA levels and PrmM-directed gene expression in SMCs. A PDGF-BB-responsive region (PRR) was also identified within PrmM, overlapping with the previously identified URR, where either siRNA knockdown of Klf4 or the combined disruption of the Klf4 elements completely abolished PDGF-BB-mediated repression of PrmM-directed gene expression in SMCs. Moreover, ChIP analysis established that PDGF-BB-induced repression of Myocd gene expression is most likely regulated by enhanced binding of Klf4 and Klf5 to a lesser extent, to the PRR of PrmM. Taken together, these data provide critical insights into the transcriptional regulation of the Myocd gene in vascular SMCs, including during SMC differentiation.
Collapse
Affiliation(s)
- Elizebeth C Turner
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland.
| | | | | | | |
Collapse
|
42
|
Sasi W, Ye L, Jiang WG, Mokbel K, Sharma A. Observations on the effects of Suppressor of Cytokine Signaling 7 (SOCS7) knockdown in breast cancer cells: their in vitro response to Insulin Like Growth Factor I (IGF-I). Clin Transl Oncol 2013; 16:476-87. [PMID: 24046004 DOI: 10.1007/s12094-013-1107-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 08/21/2013] [Indexed: 02/01/2023]
Abstract
PURPOSE Suppressor of cytokine signaling 7 (SOCS7) is a member of the SOCS family and is known to interact with phospholipase Cγ-1 (PLCγ-1), one of the insulin-like growth factor-I (IGF-I) receptor downstream molecules. In this study, we sought to observe the effect of knocking down SOCS7 gene on breast cancer cells in vitro growth and migration and to elucidate whether this involves IGF-I-PLCγ1 route using the PLCγ-1 blocker U73122. METHODS Suitable breast cancer cells (MCF7 and MDA-MB-231) were transfected with anti-SOCS7 ribozymal transgene, to create sub-lines with SOCS7 knockdown verified by RT-PCR. The growth and migration of the cells were evaluated in the presence or absence of IGF-I and PLCγ-1 inhibitor using growth assay, scratch-wound and electrical cell impedance sensing (ECIS) migration assays. RESULTS IGF-I treatment produced more pronounced influence on MCF7 growth and migration and on MDA-MB-231 migration when SOCS7 gene was knocked down in both lines (p < 0.05). The absence of IGF-I-induced growth response in MDA-MB-231 could be due to the intrinsic characteristics of these cells. PLCγ-1 pharmacological inhibition during their in vitro migration seemed to only occur when SOCS7 gene was knocked down. CONCLUSIONS To the best of our knowledge, this is the first report of the SOCS7 regulatory role in IGF-I induced in vitro functions in ER-positive and ER-negative breast cancer cells. IGF-I treatment and SOCS7 loss have synergistically resulted in increased growth and migration of MCF7 and in increased migration of MDA-MB-231 cells. The migratory effects could be due to a precise anti-PLCγ-1 role.
Collapse
Affiliation(s)
- W Sasi
- St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK,
| | | | | | | | | |
Collapse
|
43
|
Yang YR, Follo MY, Cocco L, Suh PG. The physiological roles of primary phospholipase C. Adv Biol Regul 2013; 53:232-241. [PMID: 24041464 DOI: 10.1016/j.jbior.2013.08.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/10/2013] [Indexed: 06/02/2023]
Abstract
The roles of phosphoinositide-specific phospholipase C (PLC) have been extensively investigated in diverse cell lines and pathological conditions. Among the PLC isozmes, primary PLCs, PLC-β and PLC-γ, are directly activated by receptor activation, unlike other secondary PLCs (PLC-ɛ, PLC-δ1, and PLC-η1). PLC-β isozymes are activated by G protein couple receptor and PLC-γ isozymes are activated by receptor tyrosine kinase (RTK). Primary PLCs are differentially expressed in different tissues, suggesting their specific roles in diverse tissues and regulate a variety of physiological and pathophysiological functions. Thus, dysregulation of phospholipases contributes to a number of human diseases and primary PLCs have been identified as therapeutic targets for prevention and treatment of diseases. Here we review the roles of primary PLCs in physiology and their impact in pathology.
Collapse
Affiliation(s)
- Yong Ryoul Yang
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea
| | | | | | | |
Collapse
|
44
|
Abstract
The rising epidemic of T2DM (Type 2 diabetes mellitus) worldwide is of significant concern. The inherently silent nature of the disease in its early stages precludes early detection; hence cardiovascular disease is often established by the time diabetes is diagnosed. This increased cardiovascular risk leads to significant morbidity and mortality in these individuals. Progressive development of complications as a result of previous exposure to metabolic disturbances appears to leave a long-lasting impression on cells of the vasculature that is not easily reversed and is termed 'metabolic memory'. SMCs (smooth muscle cells) of blood vessel walls, through their inherent ability to switch between a contractile quiescent phenotype and an active secretory state, maintain vascular homoeostasis in health and development. This plasticity also confers SMCs with the essential capacity to adapt and remodel in pathological states. Emerging clinical and experimental studies propose that SMCs in diabetes may be functionally impaired and thus contribute to the increased incidence of macrovascular complications. Although this idea has general support, the underlying molecular mechanisms are currently unknown and hence are the subject of intense research. The aim of the present review is to explore and evaluate the current literature relating to the problem of vascular disease in T2DM and to discuss the critical role of SMCs in vascular remodelling. Possibilities for therapeutic strategies specifically at the level of T2DM SMCs, including recent novel advances in the areas of microRNAs and epigenetics, will be evaluated. Since restoring glucose control in diabetic patients has limited effect in ameliorating their cardiovascular risk, discovering alternative strategies that restrict or reverse disease progression is vital. Current research in this area will be discussed.
Collapse
|
45
|
Chen YF, Wu KJ, Wood WG. Paeonia lactiflora Extract Attenuating Cerebral Ischemia and Arterial Intimal Hyperplasia Is Mediated by Paeoniflorin via Modulation of VSMC Migration and Ras/MEK/ERK Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:482428. [PMID: 23818926 PMCID: PMC3684030 DOI: 10.1155/2013/482428] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/10/2013] [Accepted: 05/15/2013] [Indexed: 01/21/2023]
Abstract
Paeonia lactiflora is a well-known traditional Chinese medicine. Paeoniflorin is an active component found in Paeonia lactiflora, which is used to treat smooth muscle spasms and pain and to protect the cardiovascular system. The objective of this study was to determine if Paeonia lactiflora would be protective in rodent models of cerebral ischemia and arterial intimal hyperplasia. Paeonia lactiflora extract (PLex) and paeoniflorin (PF) significantly attenuated cerebral infarction in ischemia/reperfusion injury rats and the severity of intimal hyperplasia in mice where the carotid artery was ligated. PLex and PF reduced PDGF-stimulated VSMC proliferation and migration in a dose-dependent manner by MTT, wound healing, and transwell assays. PF significantly reduced protein levels of Ras, MEK, p-MEK and p-ERK, but not MMP-2 and MMP-9. In summary, Paeonia lactiflora reduced cerebral ischemia and arterial intimal hyperplasia which were mainly made via the intermediary of PF. The protective effect of PF was related to the modulation of the Ras/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Yuh-Fung Chen
- Department of Pharmacology, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan
- Department of Pharmacy, China Medical University Hospital, No. 2 Yu-Der Road, Taichung 40447, Taiwan
| | - Kuo-Jen Wu
- Department of Pharmacology, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - W. Gibson Wood
- Department of Pharmacology, University of Minnesota and Geriatric Research, Education and Clinical Center, VA Medical Center, Minneapolis, MN 55455, USA
| |
Collapse
|
46
|
Dehydroglyasperin C, a component of liquorice, attenuates proliferation and migration induced by platelet-derived growth factor in human arterial smooth muscle cells. Br J Nutr 2013; 110:391-400. [PMID: 23298457 DOI: 10.1017/s0007114512005399] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Liquorice is one of the botanicals used frequently as a traditional medicine in the West and in the East. Platelet-derived growth factor (PDGF)-BB is involved in the development of CVD by inducing abnormal proliferation and migration of vascular smooth muscle cells. In our preliminary study, dehydroglyasperin C (DGC), an active compound of liquorice, showed strong antioxidant activity. Since phytochemicals with antioxidant activities showed beneficial effects on chronic inflammatory diseases, the present study aimed to investigate the effects of DGC on PDGF-induced proliferation and migration of human aortic smooth muscle cells (HASMC). Treatment of HASMC with DGC for 24 h significantly decreased PDGF-induced cell number and DNA synthesis in a dose-dependent manner without any cytotoxicity, as demonstrated by the 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide test and thymidine incorporation. Upon cell cycle analysis, DGC blocked the PDGF-induced progression through the G0/G1 to S phase of the cell cycle, and down-regulated the expression of cyclin-dependent kinase (CDK); 2, cyclin E, CDK4 and cyclin D1. Furthermore, DGC significantly attenuated PDGF-stimulated phosphorylation of PDGF receptor-b, phospholipase C-g1, AKT and extracellular-regulated kinase 1/2, and DGC inhibited cell migration and the dissociation of actin filaments by PDGF. In a rat vascular balloon injury model, DGC suppressed an excessive reduction in luminal diameters and neointimal formation compared with the control group. These results demonstrate the mechanistic basis for the prevention of CVD and the potential therapeutic properties of DGC.
Collapse
|
47
|
Nelumbo nucifera leaf extract inhibits neointimal hyperplasia through modulation of smooth muscle cell proliferation and migration. Nutrition 2013; 29:268-75. [DOI: 10.1016/j.nut.2012.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 03/22/2012] [Accepted: 04/29/2012] [Indexed: 11/21/2022]
|
48
|
Nagel MA, Traktinskiy I, Stenmark KR, Frid MG, Choe A, Gilden D. Varicella-zoster virus vasculopathy: immune characteristics of virus-infected arteries. Neurology 2012; 80:62-8. [PMID: 23243076 DOI: 10.1212/wnl.0b013e31827b1ab9] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Pathologic changes in varicella-zoster virus (VZV)-infected arteries include inflammation, thickened intima, and paucity of smooth muscle cells. Since no criteria have been established for early vs late VZV vasculopathy, we examined inflammatory cells and their distribution in 6 normal arteries, and 2 VZV-infected arteries 3 days after onset of disease (early) and 10 months after protracted neurologic disease (late). METHODS VZV-infected temporal artery obtained 3 days after onset of ischemic optic neuropathy from an 80-year-old man, VZV-infected middle cerebral artery (MCA) obtained 10 months after protracted disease from a 73-year-old man, and 5 MCAs and 1 temporal artery from normal subjects, age 22-60 years, were examined histologically and immunohistochemically using antibodies against VZV and inflammatory cell subsets. RESULTS In both early and late VZV vasculopathy, T cells, activated macrophages, and rare B cells were found in adventitia and intima. In adventitia of early VZV vasculopathy, neutrophils and VZV antigen were abundant and a thickened intima was associated with inflammatory cells in vaso vasorum vessels. In media of late VZV vasculopathy, viral antigen, but not leukocytes, was found. VZV was not seen in inflammatory cells. Inflammatory cells were absent in control arteries. CONCLUSIONS Both VZV and neutrophils exclusively in adventitia in early VZV vasculopathy indicate that disease begins there. Late VZV vasculopathy is distinguished by viral antigen without inflammation in media, revealing a human virus in an immunoprivileged arterial media. Association of thickened intima and inflammation in vaso vasorum vessels in early VZV vasculopathy support the role of virus-induced inflammation in vessel wall remodeling.
Collapse
Affiliation(s)
- Maria A Nagel
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Chen KC, Juo SHH. MicroRNAs in atherosclerosis. Kaohsiung J Med Sci 2012; 28:631-40. [DOI: 10.1016/j.kjms.2012.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/20/2012] [Indexed: 02/04/2023] Open
|
50
|
Contini GA, Nicolini F, Fortuna D, Pacini D, Gabbieri D, Vignali L, Valgimigli M, Manari A, Zussa C, Guastaroba P, De Palma R, Grilli R, Gherli T. Five-year outcomes of surgical or percutaneous myocardial revascularization in diabetic patients. Int J Cardiol 2012; 168:1028-33. [PMID: 23164591 DOI: 10.1016/j.ijcard.2012.10.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 10/09/2012] [Accepted: 10/28/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND The study compares five-year clinical outcomes of CABG vs PCI in a real world population of diabetic patients with multivessel coronary disease since it is not clear whether to prefer surgical or percutaneous revascularization. METHODS Between July 2002 and December 2008, 2885 multivessel coronary diabetic patients underwent revascularization (1466 CABG and 1419 PCI) at hospitals in Emilia-Romagna Region, Italy and were followed for 1827 ± 617 days by record linkage of two clinical registries with the regional administrative database of hospital admissions and the mortality registry. Five-year incidences of MACCE (mortality, acute myocardial infarction [AMI], stroke, and repeat revascularization [TVR]) were assessed with Kaplan-Meier estimates, Cox proportional hazards regression and cumulative incidence functions of death and TVR, to evaluate the competing risk of AMI on death and TVR. The same analyses were applied to the propensity score matched subgroup of patients undergoing CABG or PCI with DES and with complete revascularization. RESULTS PCI had higher mortality for all causes (HR: 1.8, 95% CI 1.4-2.2 p<0.0001), AMI (HR: 3.3, 95% CI 2.4-4.6 p<0.0001) and TVR (HR: 4.5, 95% CI 3.4-6.1 p<0.0001). No significant differences emerged for stroke (HR: 0.8, 95% CI 0.5-1.2 p=0.26). The higher incidence of AMI caused higher mortality in PCI group. Results did not change comparing CABG with PCI patients receiving complete revascularization or DES only. CONCLUSIONS Diabetics show a higher incidence of MACCE with PCI than with CABG: thus diabetes and its degree of control should be considered when choosing the type of revascularization.
Collapse
Affiliation(s)
- Giovanni Andrea Contini
- Unità Operativa di Cardiochirurgia, Dipartimento Cardio-nefro-polmonare, Azienda Ospedaliero-Universitaria di Parma, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|