1
|
Shao W, Li Z, Wang B, Gong S, Wang P, Song B, Chen Z, Feng Y. Dimethyloxalylglycine Attenuates Steroid-Associated Endothelial Progenitor Cell Impairment and Osteonecrosis of the Femoral Head by Regulating the HIF-1α Signaling Pathway. Biomedicines 2023; 11:biomedicines11040992. [PMID: 37189610 DOI: 10.3390/biomedicines11040992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 05/17/2023] Open
Abstract
Endothelial impairment and dysfunction are closely related to the pathogenesis of steroid-associated osteonecrosis of the femoral head (SONFH). Recent studies have showed that hypoxia inducible factor-1α (HIF-1α) plays a crucial role in endothelial homeostasis maintenance. Dimethyloxalylglycine (DMOG) could suppress HIF-1 degradation and result in nucleus stabilization by repressing prolyl hydroxylase domain (PHD) enzymatic activity. Our results showed that methylprednisolone (MPS) remarkably undermined biological function of endothelial progenitor cells (EPC) by inhibiting colony formation, migration, angiogenesis, and stimulating senescence of EPCs, while DMOG treatment alleviated these effects by promoting HIF-1α signaling pathway, as evidenced by senescence-associated β-galactosidase (SA-β-Gal) staining, colony-forming unit, matrigel tube formation, and transwell assays. The levels of proteins related to angiogenesis were determined by ELISA and Western blotting. In addition, active HIF-1α bolstered the targeting and homing of endogenous EPCs to the injured endothelium in the femoral head. Histopathologically, our in vivo study showed that DMOG not only alleviated glucocorticoid-induced osteonecrosis but also promoted angiogenesis and osteogenesis in the femoral head as detected by microcomputed tomography (Micro-CT) analysis and histological staining of OCN, TRAP, and Factor Ⅷ. However, all of these effects were impaired by an HIF-1α inhibitor. These findings demonstrate that targeting HIF-1α in EPCs may constitute a novel therapeutic approach for the treatment of SONFH.
Collapse
Affiliation(s)
- Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Song Gong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Beite Song
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixiang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
2
|
A BioID-Derived Proximity Interactome for SARS-CoV-2 Proteins. Viruses 2022; 14:v14030611. [PMID: 35337019 PMCID: PMC8951556 DOI: 10.3390/v14030611] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022] Open
Abstract
The novel coronavirus SARS-CoV-2 is responsible for the ongoing COVID-19 pandemic and has caused a major health and economic burden worldwide. Understanding how SARS-CoV-2 viral proteins behave in host cells can reveal underlying mechanisms of pathogenesis and assist in development of antiviral therapies. Here, the cellular impact of expressing SARS-CoV-2 viral proteins was studied by global proteomic analysis, and proximity biotinylation (BioID) was used to map the SARS-CoV-2 virus–host interactome in human lung cancer-derived cells. Functional enrichment analyses revealed previously reported and unreported cellular pathways that are associated with SARS-CoV-2 proteins. We have established a website to host the proteomic data to allow for public access and continued analysis of host–viral protein associations and whole-cell proteomes of cells expressing the viral–BioID fusion proteins. Furthermore, we identified 66 high-confidence interactions by comparing this study with previous reports, providing a strong foundation for future follow-up studies. Finally, we cross-referenced candidate interactors with the CLUE drug library to identify potential therapeutics for drug-repurposing efforts. Collectively, these studies provide a valuable resource to uncover novel SARS-CoV-2 biology and inform development of antivirals.
Collapse
|
3
|
Liu H, Li J, Jiang C, Yuan T, Ma H. Cellular communication network factor 1 (CCN1) knockdown exerts a protective effect for hepatic ischemia/reperfusion injury by deactivating the MEK/ERK pathway. Clin Res Hepatol Gastroenterol 2021; 45:101737. [PMID: 34144219 DOI: 10.1016/j.clinre.2021.101737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Hepatic ischemia/reperfusion injury (IRI) is an unsettled and intractable conundrum in clinical treatment after liver transplantation and resection. Cellular communication network factor 1 (CCN1) is upregulated in liver IRI and may play a key role in this process. The objective of this study is to investigate the regulatory mechanism of CCN1 in liver IRI, which may provide new insight into liver IRI clinical treatment. METHODS The hepatic ischemia/reperfusion model was established in male C57BL/6 mice by occlusion of vessels in the liver followed by reperfusion. The mice were transfected with two small interfering RNAs (siRNAs) against CCN1 for CCN1 knockdown. The hypoxia/reoxygenation (HR) model was established in vitro using mouse hepatic cells followed by transfection with a siRNA and treatment with an ERK activator TPA to confirm the effects of CCN1 on the MEK/ERK pathway in liver IRI. RESULTS In hepatic IRI, CCN1 was upregulated and its knockdown reduced alanine aminotransferase and aspartate transaminase levels, myeloperoxidase activity, and the levels of IL-6 and TNF-α. CCN1 downregulation alleviated inflammatory cell infiltration and apoptosis in the liver. The expressions of cleaved caspase-9, cleaved caspase-3, Bax, and CHOP were decreased with an increased Bcl-2 level after CCN1 knockdown. The phosphorylation and activation of proteins in ER stress and MEK/ERK pathway were inhibited by CCN1 knockdown. In vitro, the levels of proinflammatory cytokines, apoptosis-inducing proteins, and proteins in ER stress and MEK/ERK pathway, which were decreased by CCN1 knockdown in HR, were restored by TPA, confirming that the activation of ERK aggravated cell apoptosis after reoxygenation. CONCLUSION Overall, CCN1 knockdown may suppress the inflammation, apoptosis during hepatic IRI by reducing the MEK/ERK pathway activation, which may be a breakthrough point in clinical alleviation of hepatic IRI caused by liver transplantation and resection.
Collapse
Affiliation(s)
- Huanqiu Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ji Li
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Chengwei Jiang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Tong Yuan
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Haichun Ma
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
4
|
Xia LZ, Tao J, Chen YJ, Liang LL, Luo GF, Cai ZM, Wang Z. Factors Affecting the Re-Endothelialization of Endothelial Progenitor Cell. DNA Cell Biol 2021; 40:1009-1025. [PMID: 34061680 DOI: 10.1089/dna.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The vascular endothelium, which plays an essential role in maintaining the normal shape and function of blood vessels, is a natural barrier between the circulating blood and the vascular wall tissue. The endothelial damage can cause vascular lesions, such as atherosclerosis and restenosis. After the vascular intima injury, the body starts the endothelial repair (re-endothelialization) to inhibit the neointimal hyperplasia. Endothelial progenitor cell is the precursor of endothelial cells and plays an important role in the vascular re-endothelialization. However, re-endothelialization is inevitably affected in vivo and in vitro by factors, which can be divided into two types, namely, promotion and inhibition, and act on different links of the vascular re-endothelialization. This article reviews these factors and related mechanisms.
Collapse
Affiliation(s)
- Lin-Zhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Tao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yan-Jun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Ling-Li Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Gui-Fang Luo
- Department of Gynaecology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Ze-Min Cai
- Pediatrics Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
5
|
Yan J, Wang WB, Fan YJ, Bao H, Li N, Yao QP, Huo YL, Jiang ZL, Qi YX, Han Y. Cyclic Stretch Induces Vascular Smooth Muscle Cells to Secrete Connective Tissue Growth Factor and Promote Endothelial Progenitor Cell Differentiation and Angiogenesis. Front Cell Dev Biol 2020; 8:606989. [PMID: 33363166 PMCID: PMC7755638 DOI: 10.3389/fcell.2020.606989] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/10/2020] [Indexed: 02/05/2023] Open
Abstract
Endothelial progenitor cells (EPCs) play a vital role in endothelial repair following vascular injury by maintaining the integrity of endothelium. As EPCs home to endothelial injury sites, they may communicate with exposed vascular smooth muscle cells (VSMCs), which are subjected to cyclic stretch generated by blood flow. In this study, the synergistic effect of cyclic stretch and communication with neighboring VSMCs on EPC function during vascular repair was investigated. In vivo study revealed that EPCs adhered to the injury site and were contacted to VSMCs in the Sprague-Dawley (SD) rat carotid artery injury model. In vitro, EPCs were cocultured with VSMCs, which were exposed to cyclic stretch at a magnitude of 5% (which mimics physiological stretch) and a constant frequency of 1.25 Hz for 12 h. The results indicated that stretched VSMCs modulated EPC differentiation into mature endothelial cells (ECs) and promoted angiogenesis. Meanwhile, cyclic stretch upregulated the mRNA expression and secretion level of connective tissue growth factor (CTGF) in VSMCs. Recombinant CTGF (r-CTGF) treatment promoted endothelial differentiation of EPCs and angiogenesis, and increased their protein levels of FZD8 and β-catenin. CTGF knockdown in VSMCs inhibited cyclic stretch-induced EPC differentiation into ECs and attenuated EPC tube formation via modulation of the FZD8/β-catenin signaling pathway. FZD8 knockdown repressed endothelial differentiation of EPCs and their angiogenic activity. Wnt signaling inhibitor decreased the endothelial differentiation and angiogenetic ability of EPCs cocultured with stretched VSMCs. Consistently, an in vivo Matrigel plug assay demonstrated that r-CTGF-treated EPCs exhibited enhanced angiogenesis; similarly, stretched VSMCs also induced cocultured EPC differentiation toward ECs. In a rat vascular injury model, r-CTGF improved EPC reendothelialization capacity. The present results indicate that cyclic stretch induces VSMC-derived CTGF secretion, which, in turn, activates FZD8 and β-catenin to promote both differentiation of cocultured EPCs into the EC lineage and angiogenesis, suggesting that CTGF acts as a key intercellular mediator and a potential therapeutic target for vascular repair.
Collapse
Affiliation(s)
- Jing Yan
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wen-Bin Wang
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yang-Jing Fan
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Han Bao
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Na Li
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qing-Ping Yao
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Long Huo
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zong-Lai Jiang
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Han
- School of Life Sciences and Biotechnology, Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Ren J, Zhou T, Pilli VSS, Phan N, Wang Q, Gupta K, Liu Z, Sheibani N, Liu B. Novel Paracrine Functions of Smooth Muscle Cells in Supporting Endothelial Regeneration Following Arterial Injury. Circ Res 2020; 124:1253-1265. [PMID: 30739581 DOI: 10.1161/circresaha.118.314567] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Regeneration of denuded or injured endothelium is an important component of vascular injury response. Cell-cell communication between endothelial cells and smooth muscle cells (SMCs) plays a critical role not only in vascular homeostasis but also in disease. We have previously demonstrated that PKCδ (protein kinase C-delta) regulates multiple components of vascular injury response including apoptosis of SMCs and production of chemokines, thus is an attractive candidate for a role in SMC-endothelial cells communication. OBJECTIVE To test whether PKCδ-mediated paracrine functions of SMCs influence reendothelialization in rodent models of arterial injury. METHODS AND RESULTS Femoral artery wire injury was performed in SMC-conditional Prkcd knockout mice, and carotid angioplasty was conducted in rats receiving transient Prkcd knockdown or overexpression. SMC-specific knockout of Prkcd impaired reendothelialization, reflected by a smaller Evans blue-excluding area in the knockout compared with the wild-type controls. A similar impediment to reendothelialization was observed in rats with SMC-specific knockdown of Prkcd. In contrast, SMC-specific gene transfer of Prkcd accelerated reendothelialization. In vitro, medium conditioned by AdPKCδ-infected SMCs increased endothelial wound closure without affecting their proliferation. A polymerase chain reaction-based array analysis identified Cxcl1 and Cxcl7 among others as PKCδ-mediated chemokines produced by SMCs. Mechanistically, we postulated that PKCδ regulates Cxcl7 expression through STAT3 (signal transducer and activator of transcription 3) as knockdown of STAT3 abolished Cxcl7 expression. The role of CXCL7 in SMC-endothelial cells communication was demonstrated by blocking CXCL7 or its receptor CXCR2, both significantly inhibited endothelial wound closure. Furthermore, insertion of a Cxcl7 cDNA in the lentiviral vector that carries a Prkcd shRNA overcame the adverse effects of Prkcd knockdown on reendothelialization. CONCLUSIONS SMCs promote reendothelialization in a PKCδ-dependent paracrine mechanism, likely through CXCL7-mediated recruitment of endothelial cells from uninjured endothelium.
Collapse
Affiliation(s)
- Jun Ren
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Ting Zhou
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Vijaya Satish Sekhar Pilli
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Noel Phan
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Qiwei Wang
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Kartik Gupta
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Zhenjie Liu
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.).,Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University (Z.L.)
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison (N.S.)
| | - Bo Liu
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| |
Collapse
|
7
|
Li W, Du D, Li Y. Id-1 Promotes Reendothelialization In The Early Phase After Vascular Injury Through Activation Of NFkB/survivin Signaling Pathway. Drug Des Devel Ther 2019; 13:3799-3811. [PMID: 31802852 PMCID: PMC6827526 DOI: 10.2147/dddt.s208707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/02/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Percutaneous coronary intervention (PCI) treatment can benefit patients, but also cause irreversible mechanical damage to the vascular endothelium, ultimately leading to restenosis of the target vessel. Thus, achieving rapid re-endothelialization and restoring the integrity of the vascular endothelium and function plays an important role in inhibiting neointimal hyperplasia and preventing restenosis. Id1 (inhibitor of DNA binding/differentiation factor 1) plays an important role in promoting cell proliferation and angiogenesis. STUDY OBJECTIVE This study aims to investigate the relationship between Id1 and NFκB/survivin signaling pathways and their role in injured vascular repair by establishing a rat carotid balloon injury model. METHODS The carotid artery model of rat balloon injury was established. The injured common carotid artery was obtained at different time points after vascular injury. RNA and protein were extracted and the mRNA and protein expression levels of Id1, NFκB and survivin were detected in vascular injury. The NFκB blocker BAY 11-7082 and survivin blocker YM155 were used and the effects of Id1, NFκB, survivin mRNA and protein expression, revascularization of blood vessels and neointimal responsiveness after vascular injury were observed in the vascular tissues of Ad-Id1 transfected balloon injury. RESULTS Id1, NFκB and survivin were expressed in injured rat carotid arteries. Overexpression of Id1 promoted re-endothelialization of injured vessels through NFκB/survivin signaling pathway, inhibited early vascular endometrial reactive hyperplasia; blocked NFκB the/survivin signaling pathway attenuates the re-endothelialization of Ad-Id1 and the early endothelium of Ad-Id1. Blocking the NFκB/survivin signaling pathway attenuates the re-endothelialization and early reactive hyperplasia of vascular intima of Ad-Id1. CONCLUSION NF-kappa B/survivin signaling pathway may play an important role in Id1 promoting vascular re-endothelialization, inhibiting neointimal hyperplasia and preventing vascular restenosis.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiology, Beijing100017, People’s Republic of China
| | - Dayong Du
- Department of Cardiology, Beijing100017, People’s Republic of China
| | - Yuntian Li
- Department of Cardiology, Beijing100017, People’s Republic of China
| |
Collapse
|
8
|
Oh SY, Choi DH, Jin YM, Yu Y, Kim HY, Kim G, Park YS, Jo I. Optimization of Microenvironments Inducing Differentiation of Tonsil-Derived Mesenchymal Stem Cells into Endothelial Cell-Like Cells. Tissue Eng Regen Med 2019; 16:631-643. [PMID: 31824825 DOI: 10.1007/s13770-019-00221-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/04/2019] [Accepted: 09/17/2019] [Indexed: 12/24/2022] Open
Abstract
Background Stem cell engineering is appealing consideration for regenerating damaged endothelial cells (ECs) because stem cells can differentiate into EC-like cells. In this study, we demonstrate that tonsil-derived mesenchymal stem cells (TMSCs) can differentiate into EC-like cells under optimal physiochemical microenvironments. Methods TMSCs were preconditioned with Dulbecco's Modified Eagle Medium (DMEM) or EC growth medium (EGM) for 4 days and then replating them on Matrigel to observe the formation of a capillary-like network under light microscope. Microarray, quantitative real time polymerase chain reaction, Western blotting and immunofluorescence analyses were used to evaluate the expression of gene and protein of EC-related markers. Results Preconditioning TMSCs in EGM for 4 days and then replating them on Matrigel induced the formation of a capillary-like network in 3 h, but TMSCs preconditioned with DMEM did not form such a network. Genome analyses confirmed that EGM preconditioning significantly affected the expression of genes related to angiogenesis, blood vessel morphogenesis and development, and vascular development. Western blot analyses revealed that EGM preconditioning with gelatin coating induced the expression of endothelial nitric oxide synthase (eNOS), a mature EC-specific marker, as well as phosphorylated Akt at serine 473, a signaling molecule related to eNOS activation. Gelatin-coating during EGM preconditioning further enhanced the stability of the capillary-like network, and also resulted in the network more closely resembled to those observed in human umbilical vein endothelial cells. Conclusion This study suggests that under specific conditions, i.e., EGM preconditioning with gelatin coating for 4 days followed by Matrigel, TMSCs could be a source of generating endothelial cells for treating vascular dysfunction.
Collapse
Affiliation(s)
- Se-Young Oh
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Da Hyeon Choi
- 3School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Chungdae-ro 1, Seowon-Gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Yoon Mi Jin
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Yeonsil Yu
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Ha Yeong Kim
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,4Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Gyungah Kim
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Yoon Shin Park
- 3School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Chungdae-ro 1, Seowon-Gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Inho Jo
- 1Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea.,2Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| |
Collapse
|
9
|
Liu C, Cao Y, He X, Zhang C, Liu J, Zhang L, Wu D, Zhuang X, Xue R, Huang H, Jiang J, Dong B, Sun Y, Dong Y, Zhao J. Association of Cyr61-cysteine-rich protein 61 and short-term mortality in patients with acute heart failure and coronary heart disease. Biomark Med 2019; 13:1589-1597. [PMID: 31660756 DOI: 10.2217/bmm-2019-0111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aim: The protein CCN1/CYR61 exerts critical functions in myocardial ischemic injury. We sought to investigate the prognostic value of CCN1 in patients with acute heart failure (AHF) and coronary heart disease (CAD). Methodology: We prospectively enrolled 113 patients with AHF and CAD. Patients were followed for all-cause mortality during a 30-day follow-up. Logistic models were used to estimate the association of CCN1 concentrations with 30-day mortality. Results: In multivariate logistic regression model, CCN1 was a significant predictor of 30-day mortality independent of current markers. Enhanced Feedback for Effective Cardiac Treatment risk score was recommended as one of the selected multivariable risk scores to predict outcome in AHF. CCN1 improved risk stratification for all-cause mortality when added to the Enhanced Feedback for Effective Cardiac Treatment risk scores at 30 days. Conclusion: We found CCN1 is independently associated with 30-day mortality in patients with AHF and CAD.
Collapse
Affiliation(s)
- Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Yalin Cao
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550001, PR China
| | - Xin He
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Chongyu Zhang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Jian Liu
- Department of Anesthesiology, Southern Medical University Nanfang Hospital, Guangzhou 510515, PR China
| | - Lili Zhang
- Department of Cardiology, Hainan General Hospital, Haikou 570311, PR China
| | - Dexi Wu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Xiaodong Zhuang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Ruicong Xue
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Huiling Huang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Jingzhou Jiang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Bin Dong
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Yu Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Yugang Dong
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| | - Jingjing Zhao
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou 510080, PR China
| |
Collapse
|
10
|
Hu A, Huang J, Li S, Gao Y, Wu L, Deng J, Liu J, Gong Q, Li L, Xu S. Involvement of stromal cell-derived factor-1α (SDF-1α), stem cell factor (SCF), fractalkine (FKN) and VEGF in TSG protection against intimal hyperplasia in rat balloon injury. Biomed Pharmacother 2019; 110:887-894. [DOI: 10.1016/j.biopha.2018.12.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 01/17/2023] Open
|
11
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
12
|
Lu W, Li X. Vascular stem/progenitor cells: functions and signaling pathways. Cell Mol Life Sci 2018; 75:859-869. [PMID: 28956069 PMCID: PMC11105279 DOI: 10.1007/s00018-017-2662-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/05/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Vascular stem/progenitor cells (VSCs) are an important source of all types of vascular cells needed to build, maintain, repair, and remodel blood vessels. VSCs, therefore, play critical roles in the development, normal physiology, and pathophysiology of numerous diseases. There are four major types of VSCs, including endothelial progenitor cells (EPCs), smooth muscle progenitor cells (SMPCs), pericytes, and mesenchymal stem cells (MSCs). VSCs can be found in bone marrow, circulating blood, vessel walls, and other extravascular tissues. During the past two decades, considerable progress has been achieved in the understanding of the derivation, surface markers, and differentiation of VSCs. Yet, the mechanisms regulating their functions and maintenance under normal and pathological conditions, such as in eye diseases, remain to be further elucidated. Owing to the essential roles of blood vessels in human tissues and organs, understanding the functional properties and the underlying molecular basis of VSCs is of critical importance for both basic and translational research.
Collapse
Affiliation(s)
- Weisi Lu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Xuri Li
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
13
|
Extracellular Matrix, a Hard Player in Angiogenesis. Int J Mol Sci 2016; 17:ijms17111822. [PMID: 27809279 PMCID: PMC5133823 DOI: 10.3390/ijms17111822] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins, glycoproteins, proteoglycans, and polysaccharides. Through multiple interactions with each other and the cell surface receptors, not only the ECM determines the physical and mechanical properties of the tissues, but also profoundly influences cell behavior and many physiological and pathological processes. One of the functions that have been extensively explored is its impingement on angiogenesis. The strong impact of the ECM in this context is both direct and indirect by virtue of its ability to interact and/or store several growth factors and cytokines. The aim of this review is to provide some examples of the complex molecular mechanisms that are elicited by these molecules in promoting or weakening the angiogenic processes. The scenario is intricate, since matrix remodeling often generates fragments displaying opposite effects compared to those exerted by the whole molecules. Thus, the balance will tilt towards angiogenesis or angiostasis depending on the relative expression of pro- or anti-angiogenetic molecules/fragments composing the matrix of a given tissue. One of the vital aspects of this field of research is that, for its endogenous nature, the ECM can be viewed as a reservoir to draw from for the development of new more efficacious therapies to treat angiogenesis-dependent pathologies.
Collapse
|
14
|
Wang H, Yin YG, Huang H, Zhao XH, Yu J, Wang Q, Li W, Cai KY, Ding SF. Transplantation of EPCs overexpressing PDGFR-β promotes vascular repair in the early phase after vascular injury. BMC Cardiovasc Disord 2016; 16:179. [PMID: 27619504 PMCID: PMC5020463 DOI: 10.1186/s12872-016-0353-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/26/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) play important roles in the regeneration of the vascular endothelial cells (ECs). Platelet-derived growth factor receptor (PDGFR)-β is known to contribute to proliferation, migration, and angiogenesis of EPCs, this study aims to investigate effects of transplantation of EPCs overexpressing PDGFR-β on vascular regeneration. METHODS We transplanted genetically modified EPCs overexpressing PDGFR-β into a mouse model with carotid artery injury. After 3 days of EPCs transplantation, the enhanced green fluorescent protein (EGFP)-expressing cells were found at the injury site and the lining of the lumen by laser scanning confocal microscope (LSCM). At 4, 7, and 14 days of the carotid artery injury, reendothelialization was evaluated by Evans Blue staining. Neointima formation was evaluated at day 14 with hematoxylin and eosin (HE) staining by calculating the neointimal area, medial area, and neointimal/media (NI/M) ratio. Intimal cell apoptosis was evaluated using TUNEL assay. Then we tested whether PDGF-BB-induced VSMC migration and PDGF-BB's function in reducing VSMC apoptosis can be attenuated by EPCs overexpressing PDGFR-β in a transwell co-culture system. RESULTS Our results showed that EPCs overexpressing PDGFR-β accelerates reendothelialization and mitigates neointimal formation at 14 days after injury. Moreover, we found that there is great possibility that EPCs overexpressing PDGFR-β enhanc VSMC apoptosis and suppress VSMC migration by competitive consumption of PDGF-BB in the early phase after carotid artery injury in mice. CONCLUSIONS We report the first in vivo and in vitro evidence that transplantation of genetically modified EPC can have a combined effect of both amplifying the reendothelialization capacity of EPCs and inhibiting neointima formation so as to facilitate better inhibition of adverse remodeling after vascular injury.
Collapse
Affiliation(s)
- Hang Wang
- Cadre Ward Two, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China
| | - Yang-Guang Yin
- Intensive Care Unit, The sixth people's hospital of Chongqing, Nan'an District, Chongqing, 400060, China
| | - Hao Huang
- Clinic center, Shenzhen Hornetcorn Biotechnology Company, Ltd, Shenzhen, 518400, China
| | - Xiao-Hui Zhao
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jie Yu
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qiang Wang
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Wei Li
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ke-Yin Cai
- Cadre Ward Two, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China
| | - Shi-Fang Ding
- Institute of Cardiovascular Science, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China.
| |
Collapse
|
15
|
Sinning C, Schnabel RB, Zeller T, Seiffert M, Rupprecht HJ, Lackner KJ, Blankenberg S, Bickel C, Westermann D. Prognostic use of soluble fms-like tyrosine kinase-1 and placental growth factor in patients with coronary artery disease. Biomark Med 2016; 10:95-106. [DOI: 10.2217/bmm.15.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Intention of the study is to assess the cardiovascular mortality of patients with coronary artery disease (CAD) with the biomarkers of angiogenesis PlGF and its endogenous inhibitor sFlt-1. Methods: The cohort included n = 1848 patients with CAD and 282 subjects without CAD. In 85 patients cardiovascular mortality, as combination of fatal myocardial infarction or any cardiac death, during a median follow-up duration of 3.9 years was reported. Results: In Kaplan–Meier curve analysis PlGF in rising thirds was not predictive regarding outcome (p = 0.54), the same was shown for sFlt-1 (p = 0.44). Cox regression for the fully adjusted model provided a hazard ratio (HR) of 0.8 (p = 0.18) for PlGF and for sFlt-1 a HR = 1.0 (p = 0.8). Conclusion: Our results point out that these biomarkers reflecting angiogenesis might not be suited to establish prognosis in CAD.
Collapse
Affiliation(s)
- Christoph Sinning
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Renate B Schnabel
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Tanja Zeller
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Moritz Seiffert
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Hans J Rupprecht
- Department of Internal Medicine II, GPR Klinikum Rüsselsheim, Germany
| | - Karl J Lackner
- Department of Clinical Chemistry & Laboratory Medicine, Johannes Gu-tenberg-University Mainz, Germany
| | - Stefan Blankenberg
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Christoph Bickel
- Department of Internal Medicine, Federal Armed Forces Central Hospital, Koblenz, Germany
| | - Dirk Westermann
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| |
Collapse
|
16
|
Wu X, Yin T, Tian J, Tang C, Huang J, Zhao Y, Zhang X, Deng X, Fan Y, Yu D, Wang G. Distinctive effects of CD34- and CD133-specific antibody-coated stents on re-endothelialization and in-stent restenosis at the early phase of vascular injury. Regen Biomater 2015; 2:87-96. [PMID: 26813006 PMCID: PMC4669017 DOI: 10.1093/rb/rbv007] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 01/01/2023] Open
Abstract
It is not clear what effects of CD34- and CD133-specific antibody-coated stents have on re-endothelialization and in-stent restenosis (ISR) at the early phase of vascular injury. This study aims at determining the capabilities of different coatings on stents (e.g. gelatin, anti-CD133 and anti-CD34 antibodies) to promote adhesion and proliferation of endothelial progenitor cells (EPCs). The in vitro study revealed that the adhesion force enabled the EPCs coated on glass slides to withstand flow-induced shear stress, so that allowing for the growth of the cells on the slides for 48 h. The in vivo experiment using a rabbit model in which the coated stents with different substrates were implanted showed that anti-CD34 and anti-CD133 antibody-coated stents markedly reduced the intima area and restenosis than bare mental stents (BMS) and gelatin-coated stents. Compared with the anti-CD34 antibody-coated stents, the time of cells adhesion was longer and earlier present in the anti-CD133 antibody-coated stents and anti-CD133 antibody-coated stents have superiority in re-endothelialization and inhibition of ISR. In conclusion, this study demonstrated that anti-CD133 antibody as a stent coating for capturing EPCs is better than anti-CD34 antibody in promoting endothelialization and reducing ISR.
Collapse
Affiliation(s)
- Xue Wu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Tieying Yin
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Jie Tian
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Chaojun Tang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Junli Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Yinping Zhao
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Xiaojuan Zhang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Xiaoyan Deng
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Yubo Fan
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Donghong Yu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College, Chongqing University, No. 174 Shazhengjie, Shapingba, Chongqing 400044, China, School of Biological Science and Medical Engineering, Beihang University, No. 37 XueYuan Road, HaiDian District, Beijing 100191, China and Department of Biotechnology, Chemistry, and Environmental Engineering, Aalborg University, Fredrik Bajers Vej 7, Building H, 9220, Aalborg, Denmark
| |
Collapse
|
17
|
Goh ET, Wong E, Farhatnia Y, Tan A, Seifalian AM. Accelerating in situ endothelialisation of cardiovascular bypass grafts. Int J Mol Sci 2014; 16:597-627. [PMID: 25551605 PMCID: PMC4307264 DOI: 10.3390/ijms16010597] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 12/19/2014] [Indexed: 12/18/2022] Open
Abstract
The patency of synthetic cardiovascular grafts in the long run is synonymous with their ability to inhibit the processes of intimal hyperplasia, thrombosis and calcification. In the human body, the endothelium of blood vessels exhibits characteristics that inhibit such processes. As such it is not surprising that research in tissue engineering is directed towards replicating the functionality of the natural endothelium in cardiovascular grafts. This can be done either by seeding the endothelium within the lumen of the grafts prior to implantation or by designing the graft such that in situ endothelialisation takes place after implantation. Due to certain difficulties identified with in vitro endothelialisation, in situ endothelialisation, which will be the focus of this article, has garnered interest in the last years. To promote in situ endothelialisation, the following aspects can be taken into account: (1) Endothelial progenital cell mobilization, adhesion and proliferation; (2) Regulating differentiation of progenitor cells to mature endothelium; (3) Preventing thrombogenesis and inflammation during endothelialisation. This article aims to review and compile recent developments to promote the in situ endothelialisation of cardiovascular grafts and subsequently improve their patency, which can also have widespread implications in the field of tissue engineering.
Collapse
Affiliation(s)
- Ee Teng Goh
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Eleanor Wong
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Yasmin Farhatnia
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Aaron Tan
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Alexander M Seifalian
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| |
Collapse
|
18
|
Hviid CVB, Samulin Erdem J, Drechsler S, Weixelbaumer K, Ahmed MS, Attramadal H, Redl H, Bahrami S, Osuchowski MF, Aasen AO. The matricellular "cysteine-rich protein 61" is released from activated platelets and increased in the circulation during experimentally induced sepsis. Shock 2014; 41:233-40. [PMID: 24430538 DOI: 10.1097/shk.0000000000000105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sepsis and sepsis-induced organ dysfunction remain lethal and common conditions among intensive care patients. Accumulating evidence suggests that the matricellular Cyr61/CCN1 (cysteine-rich, angiogenic-inducer, 61) protein is involved in the regulation of inflammatory responses and possesses organ-protective capabilities in diseases of an inflammatory etiology. However, its regulation in sepsis remains largely unexplored. The present study provides a comprehensive description of CCN1 regulation in the circulation and vital organs during experimentally induced sepsis with developing organ dysfunction. Female CD-1 mice served as baseline controls or were subjected to cecal ligation and puncture (CLP) for 18 to 96 h, and CCN1 regulation was analyzed in selected organs and in the circulation. A 5-, 5-, and 3-fold increases in circulating CCN1 protein were observed at 18, 48, and 96 h after CLP, respectively. Hepatic and pulmonary CCN1 mRNA expression was down-regulated by 80%, 60%, and 55% and 85%, 80%, and 65% at 18, 48, and 96 h after CLP and undetectable in circulating white blood cells. To identify a potential source for the circulating protein, mouse and human platelets were explored and revealed to contain CCN1. Human platelets were stimulated by thrombin and a specific PAR1 agonist (SFLLRN) in vitro. Both agonists induced an instant CCN1 release, and the effect of SFLLRN was blocked by the specific antagonist RWJ56110. The current study demonstrates that experimental sepsis is associated with a robust increase in circulating CCN1 protein levels and a paradoxical downregulation of CCN1 mRNA expression in vital organs. It provides evidence that CCN1 is released from activated platelets, suggesting that platelets constitute a novel source for CCN1 release to the circulation during sepsis.
Collapse
Affiliation(s)
- Claus Vinter B Hviid
- *Institute for Surgical Research, Oslo University Hospital HF-Rikshospitalet; †Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and ‡Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Brenner C, Kränkel N, Kühlenthal S, Israel L, Remm F, Fischer C, Herbach N, Speer T, Grabmaier U, Laskowski A, Gross L, Theiss H, Wanke R, Landmesser U, Franz WM. Short-term inhibition of DPP-4 enhances endothelial regeneration after acute arterial injury via enhanced recruitment of circulating progenitor cells. Int J Cardiol 2014; 177:266-75. [PMID: 25499391 DOI: 10.1016/j.ijcard.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/06/2014] [Accepted: 09/15/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Endothelial injuries regularly occur in atherosclerosis and during interventional therapies of the arterial occlusive disease. Disturbances in the endothelial integrity can lead to insufficient blood supply and bear the risk of thrombus formation and acute vascular occlusion. At present, effective therapeutics to restore endothelial integrity are barely available. We analyzed the effect of pharmacological DPP-4-inhibition by Sitagliptin on endogenous progenitor cell-based endothelial regeneration via the SDF-1α/CXCR4-axis after acute endothelial damage in a mouse model of carotid injury. METHODS AND RESULTS Induction of a defined endothelial injury was performed in the carotid artery of C57Bl/6 mice which led to a local upregulation of SDF-1α expression. Animals were treated with placebo, Sitagliptin or Sitagliptin+AMD3100. Using mass spectrometry we could prove that Sitagliptin prevented cleavage of the chemokine SDF-1α. Accordingly, increased SDF-1α concentrations enhanced recruitment of systemically applied and endogenous circulating CXCR4+ progenitor cells to the site of vascular injury followed by a significantly accelerated reendothelialization as compared to placebo-treated animals. Improved endothelial recovery, as well as recruitment of circulating CXCR4+ progenitor cells (CD133+, Flk1+), was reversed by CXCR4-antagonization through AMD3100. In addition, short-term Sitagliptin treatment did not significantly promote neointimal or medial hyperplasia. CONCLUSION Sitagliptin can accelerate endothelial regeneration after acute endothelial injury. DPP-4 inhibitors prevent degradation of the chemokine SDF-1α and thus improve the recruitment of regenerative circulating CXCR4+ progenitor cells which mediate local endothelial cell proliferation without adversely affecting vessel wall architecture.
Collapse
Affiliation(s)
- Christoph Brenner
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| | - Nicolle Kränkel
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Sarah Kühlenthal
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lars Israel
- Institute of Molecular Biology, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Friederike Remm
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Cornelia Fischer
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Timo Speer
- Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Ulrich Grabmaier
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Alexandra Laskowski
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lisa Gross
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Hans Theiss
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Ulf Landmesser
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Wolfgang-Michael Franz
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
20
|
Abstract
Endothelial progenitor cells (EPCs) are primitive endothelial precursors which are known to functionally contribute to the pathogenesis of disease. To date a number of distinct subtypes of these cells have been described, with differing maturation status, cellular phenotype, and function. Although there is much debate on which subtype constitutes the true EPC population, all subtypes have endothelial characteristics and contribute to neovascularisation. Vasculogenesis, the process by which EPCs contribute to blood vessel formation, can be dysregulated in disease with overabundant vasculogenesis in the context of solid tumours, leading to tumour growth and metastasis, and conversely insufficient vasculogenesis can be present in an ischemic environment. Importantly, it is widely known that transcription factors tightly regulate cellular phenotype and function by controlling the expression of particular target genes and in turn regulating specific signalling pathways. This suggests that transcriptional regulators may be potential therapeutic targets to control EPC function. Herein, we discuss the observed EPC subtypes described in the literature and review recent studies describing the role of a number of transcriptional families in the regulation of EPC phenotype and function in normal and pathological conditions.
Collapse
|
21
|
Li W, Cao Z, Xia Z, Meng Q, Yu WM, Yao X, Cheng F. Acute kidney injury induced by various pneumoperitoneum pressures in a rabbit model of mild and severe hydronephrosis. Urol Int 2014; 94:225-33. [PMID: 25196500 DOI: 10.1159/000362845] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/14/2014] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Increased pneumoperitoneum pressure during laparoscopic surgery can result in acute kidney injury. We aimed to clarify whether intraabdominal pressure tolerance is modified in various degrees of unilateral kidney hydronephrosis in rabbits. METHODS A total 90 rabbits were randomly allocated to three groups (group PN, PM and PS, i.e. rabbits with no, mild and severe hydronephrosis, respectively, subjected to intraabdominal pressures). Rabbits in group PM (n=30) and group PS (n=30) underwent a surgical procedure inducing a mild or severe left hydronephrosis. Rabbits in all groups were then allocated to 5 subgroups. Then, they were subjected to intraabdominal pressures of 0, 6, 9, 12, and 15 mm Hg, respectively. Acute kidney injury was assessed by measuring serum creatinine (Scr), blood urea nitrogen (BUN), tubular cell apoptosis, kidney injury molecule-1 (KIM-1) and cysteine-rich 61 (Cyr-61/CCN1) expression. RESULTS Acute kidney injury with increased tubular apoptosis and KIM-1 and Cyr-61 expression occurred when intraabdominal pressure reached 15, 15 and 9 mm Hg in PN, PM and PS groups, respectively. The Scr and BUN levels were similar in all groups. CONCLUSIONS In rabbits, kidneys with severe hydronephrosis were more likely to suffer acute injury when they were exposed to pneumoperitoneal pressure.
Collapse
Affiliation(s)
- Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Yin C, Liang Y, Guo S, Zhou X, Pan X. CCN1 enhances angiogenic potency of bone marrow transplantation in a rat model of hindlimb ischemia. Mol Biol Rep 2014; 41:5813-8. [DOI: 10.1007/s11033-014-3455-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
|
23
|
Katsaros KM, Kastl SP, Krychtiuk KA, Hutter R, Zorn G, Maurer G, Huber K, Wojta J, Christ G, Speidl WS. An increase of VEGF plasma levels is associated with restenosis of drug-eluting stents. EUROINTERVENTION 2014; 10:224-30. [DOI: 10.4244/eijv10i2a36] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Cao Z, Yu W, Li W, Cheng F, Xia Y, Rao T, Yao X, Zhang X, Larré S. Acute kidney injuries induced by various irrigation pressures in rat models of mild and severe hydronephrosis. Urology 2013; 82:1453.e9-16. [PMID: 24144540 DOI: 10.1016/j.urology.2013.08.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 07/22/2013] [Accepted: 08/09/2013] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To clarify whether tolerance to irrigation pressure could be modified over varying degrees of kidney obstruction during the endoscopic treatment of kidney stones in a rat model. METHODS A total of 126 rats were randomly allocated into 2 experimental groups and a control group. The experimental groups underwent a surgical procedure to induce mild (group M, n = 60) or severe (group S, n = 60) hydronephrosis. In each group, the rats were then randomly allocated into 4 subgroups (M0 to M3 and S0 to S3) of respectively 6, 18, 18, and 18 rats. Groups 0 to 3 were respectively perfused with 0 (no irrigation), 20, 60, and 100 mm Hg pressure fluid. The control group underwent no surgical procedures and was only perfused with 100 mm Hg pressure fluid. Acute kidney injuries were assessed by analyzing the kidney microstructure, tubular cell apoptosis, kidney injury molecule-1, and cysteine-rich 61 (Cyr61/CCN1) expression using immunohistochemistry. RESULTS No abnormalities were observed for the control group, groups 0, or 1. In group 2, abnormalities were observed only in the S group, whereas all kidneys in group 3 suffered acute kidneys injuries, along with occurrence of tubular cells necrosis, increased apoptosis, and increased expression of kidney injury molecule-1 and Cyr61. CONCLUSION Rats with severely obstructed kidneys were more likely to suffer acute kidney injuries than those with less obstructed kidneys when exposed to higher kidney irrigation pressures. This suggests that the pressure should be controlled and reduced when performing endourologic procedures in the context of kidney obstruction.
Collapse
Affiliation(s)
- Zhixiu Cao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang N, Zhang R, Wang SJ, Zhang CL, Mao LB, Zhuang CY, Tang YY, Luo XG, Zhou H, Zhang TC. Vascular endothelial growth factor stimulates endothelial differentiation from mesenchymal stem cells via Rho/myocardin-related transcription factor--a signaling pathway. Int J Biochem Cell Biol 2013; 45:1447-56. [PMID: 23624342 DOI: 10.1016/j.biocel.2013.04.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/23/2013] [Accepted: 04/15/2013] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) are pluripotent progenitors that can differentiate into a variety of cell types. Vascular endothelial growth factor (VEGF) is one of the major factors of initiating and regulating angiogenesis. It has been reported that VEGF can induce MSCs differentiated into endothelial cells (ECs). However, the mechanism that VEGF-induced MSC differentiation is not completely understood. Here, we showed that VEGF induced human and rat bone marrow-derived MSCs differentiation to ECs. Rho family plays an important role in VEGF-induced endothelial cell migration and angiogenesis. Our results indicated that in MSCs, VEGF activated Rho/ROCK signaling pathway and promoted nuclear translocation of myocardin-related transcription factor-A (MRTF-A), which is controlled by Rho/ROCK signaling. In addition, Rho inhibitor C3 transferase, ROCK inhibitor Y27632 or depletion of endogenous MRTF-A abolished the VEGF-induced differentiation of MSCs into ECs. Furthermore, VEGF also enhanced the expression levels of CYR61/CCN1, as a regulator of vascular development and angiogenesis, and knockdown of endogenous MRTF-A reduced VEGF-induced the upregulation of CYR61/CCN1. Report assays with site-direct mutation analysis of CYR61/CCN1 promoter demonstrated that MRTF-A transactivated CYR61/CCN1 promoter mainly depending on CArG box. In this study, we identify the Rho/MRTF-A signaling pathway as a main actor in controlling VEGF-induced differentiation of human and rat bone marrow-derived MSCs into endothelial cells.
Collapse
Affiliation(s)
- Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ischemic preconditioning increases endothelial progenitor cell number to attenuate partial nephrectomy-induced ischemia/reperfusion injury. PLoS One 2013; 8:e55389. [PMID: 23383174 PMCID: PMC3561290 DOI: 10.1371/journal.pone.0055389] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/21/2012] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES The objective of this study was to investigate the role of endothelial progenitor cells (EPCs) in the modulation of ischemia-reperfusion injury (IRI) in a partial nephrectomy (PN) rat model using early-phase ischemic preconditioning (IPC). MATERIALS AND METHODS Ninety male Sprague-Dawley rats were randomly divided into three groups following right-side nephrectomy: Sham-operated rats (surgery without vascular clamping); PN rats (renal blood vessels were clamped for 40 min and PN was performed); and IPC rats (pretreated with 15 min ischemia and 10 min reperfusion). At 1, 3, 6, 12, 24 h, and 3 days after reperfusion, the pool of circulating EPCs and kidneys were harvested. The extent of renal injury was assessed, along with EPC number, cell proliferation, angiogenesis, and vascular growth factor expression. RESULTS Pretreated rats exhibited significant improvements in renal function and morphology. EPC numbers in the kidneys were increased at 12 h following reperfusion in the IPC group as compared to the PN or Sham groups. Cell proliferation (including endothelial and tubular epithelial cells) and angiogenesis in peritubular capillaries were markedly increased in kidneys treated with IPC. In addition, vascular endothelial growth factor-A (VEGF-A) and stromal cell-derived factor-1α (SDF-1α) expression in the kidneys of pretreated rats was increased compared to rats subjected to PN. CONCLUSIONS OUR INVESTIGATION SUGGESTED THAT: (1) the early phase of IPC may attenuate renal IRI induced by PN; (2) EPCs play an important role in renal protection, involving promotion of cell proliferation and angiogenesis through release of several angiogenic factors.
Collapse
|
27
|
Lorenzen JM, Dietrich B, Fiedler J, Jazbutyte V, Fleissner F, Karpinski N, Weidemann F, Wanner C, Asan E, Caprio M, Ertl G, Bauersachs J, Thum T. Pathologic endothelial response and impaired function of circulating angiogenic cells in patients with Fabry disease. Basic Res Cardiol 2012. [PMID: 23184391 DOI: 10.1007/s00395-012-0311-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Fabry disease is an X-chromosomal recessive deficiency of the lysosomal hydrolase alpha-galactosidase A (alpha-Gal). This results in an accumulation of globotriaosylceramide (GL-3) in a variety of cells often with subsequent functional impairment. Here, the impact of Fabry disease on the biology of circulating angiogenic cells (CACs) and the endothelial response to transient ischemia was investigated. Untreated patients with Fabry disease (n = 26), patients after initiation of alpha-Gal enzyme replacement therapy (ERT) (n = 16) and healthy controls (n = 26) were investigated. Endothelial function was assessed by the EndoPAT2000 device. CAC numbers were assessed by flow-cytometry, CAC function by a modified Boyden chamber assay. Fabry patients showed a pathologic endothelial response, which normalized after ERT. CACs were increased in number, but functionally impaired. Immunofluorescence and electron microscopy identified an accumulation of GL-3 in Fabry CACs. ERT attenuated CAC dysfunction and improved markers of oxidative stress response in Fabry patients via a reduction in GL-3 accumulation in vitro and in vivo. Silencing of alpha-Gal in healthy CACs impaired their migratory capacity underlining a key role of this enzyme for CAC function. CAC supernatant as well as CACs from Fabry patients impaired angiogenesis and migratory capacity of HUVECs providing a mechanistic link between CAC and endothelial dysfunction. CAC adhesion to TNF-α pre-stimulated HUVECs and tube formation was impaired by alpha-Gal knockdown. Fabry patients show a dysfunction of CAC and a pathologic endothelial response. ERT improves CAC and endothelial function and thus may attenuate development of cardiovascular disease in the long term in this patient population.
Collapse
Affiliation(s)
- Johan M Lorenzen
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A novel model of intimal hyperplasia with graded hypoosmotic damage. Cardiovasc Pathol 2012; 21:490-8. [DOI: 10.1016/j.carpath.2012.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/19/2012] [Accepted: 02/20/2012] [Indexed: 11/24/2022] Open
|
29
|
Szabó PM, Pintér M, Szabó DR, Zsippai A, Patócs A, Falus A, Rácz K, Igaz P. Integrative analysis of neuroblastoma and pheochromocytoma genomics data. BMC Med Genomics 2012; 5:48. [PMID: 23106811 PMCID: PMC3495658 DOI: 10.1186/1755-8794-5-48] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 10/26/2012] [Indexed: 12/26/2022] Open
Abstract
Background Pheochromocytoma and neuroblastoma are the most common neural crest-derived tumors in adults and children, respectively. We have performed a large-scale in silico analysis of altogether 1784 neuroblastoma and 531 pheochromocytoma samples to establish similarities and differences using analysis of mRNA and microRNA expression, chromosome aberrations and a novel bioinformatics analysis based on cooperative game theory. Methods Datasets obtained from Gene Expression Omnibus and ArrayExpress have been subjected to a complex bioinformatics analysis using GeneSpring, Gene Set Enrichment Analysis, Ingenuity Pathway Analysis and own software. Results Comparison of neuroblastoma and pheochromocytoma with other tumors revealed the overexpression of genes involved in development of noradrenergic cells. Among these, the significance of paired-like homeobox 2b in pheochromocytoma has not been reported previously. The analysis of similar expression patterns in neuroblastoma and pheochromocytoma revealed the same anti-apoptotic strategies in these tumors. Cancer regulation by stathmin turned out to be the major difference between pheochromocytoma and neuroblastoma. Underexpression of genes involved in neuronal cell-cell interactions was observed in unfavorable neuroblastoma. By the comparison of hypoxia- and Ras-associated pheochromocytoma, we have found that enhanced insulin like growth factor 1 signaling may be responsible for the activation of Src homology 2 domain containing transforming protein 1, the main co-factor of RET. Hypoxia induced factor 1α and vascular endothelial growth factor signaling included the most prominent gene expression changes between von Hippel-Lindau- and multiple endocrine neoplasia type 2A-associated pheochromocytoma. Conclusions These pathways include previously undescribed pathomechanisms of neuroblastoma and pheochromocytoma and associated gene products may serve as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Peter M Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str, 46, Budapest, H-1088, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
30
|
An essential role for the Id1/PI3K/Akt/NFkB/survivin signalling pathway in promoting the proliferation of endothelial progenitor cells in vitro. Mol Cell Biochem 2011; 363:135-45. [PMID: 22139302 PMCID: PMC3289789 DOI: 10.1007/s11010-011-1166-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/23/2011] [Indexed: 12/24/2022]
Abstract
The enhancement of re-endothelialisation is a critical therapeutic option for repairing injured blood vessels. Endothelial progenitor cells (EPCs) are the major source of cells that participate in endothelium repair and contribute to re-endothelialisation by reducing neointima formation after vascular injury. The over-expression of the inhibitor of differentiation or DNA binding 1 (Id1) significantly improved EPC proliferation. This study aimed to investigate the effects of Id1 on the phosphatidylinositol-3-kinase (PI3K)/Akt/nuclear factor kappa B (NFκB)/survivin signalling pathway and its significance in promoting EPC proliferation in vitro. Spleen-derived EPCs were cultured as previously described. Id1 was presented at low levels in EPCs, and was rapidly up-regulated by stimulation with vascular endothelial growth factor. We demonstrated that transient transfection of Id1 into EPCs activated the PI3K/Akt/NFκB/survivin signalling pathway and promoted EPC proliferation. The proliferation of EPCs was extensively inhibited by silencing of endogenous Id1, and knockdown of Id1 expression led to suppression of PI3K/Akt/NFκB/survivin signalling pathway in EPCs. In addition, blockade by the PI3K-specific inhibitor LY294002, Akt inhibitor, the NFκB inhibitor BAY 11-7082, the survivin inhibitor Curcumin, or the survivin inhibitor YM155 reduced the effects of Id1 transfection. These results suggest that the Id1/PI3K/Akt/NFκB/survivin signalling pathway plays a critical role in EPC proliferation. The Id1/PI3K/Akt/NFκB/survivin signalling pathway may represent a novel therapeutic target in the prevention of restenosis after vascular injury.
Collapse
|
31
|
Presence of endothelial colony-forming cells is associated with reduced microvascular obstruction limiting infarct size and left ventricular remodelling in patients with acute myocardial infarction. Basic Res Cardiol 2011; 106:1397-410. [PMID: 21904841 DOI: 10.1007/s00395-011-0220-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/04/2011] [Accepted: 08/29/2011] [Indexed: 12/24/2022]
Abstract
Endothelial colony-forming cells (ECFCs) are known to increase after acute myocardial infarction (AMI). We examined whether the presence of ECFCs is associated with preserved microvascular integrity in the myocardium at risk by reducing microvascular obstruction (MVO). We enrolled 88 patients with a first ST elevation AMI. ECFC colonies and circulating progenitor cells were characterized at admission. MVO was evaluated at 5 days and infarct size at 5 days and at 6-month follow-up by magnetic resonance imaging. ECFC colonies were detected in 40 patients (ECFC(pos) patients). At 5 days, MVO was of greater magnitude in ECFC(neg) versus ECFC(pos) patients (7.7 ± 5.3 vs. 3.2 ± 5%, p = 0.0002). At 6 months, in ECFC(pos) patients, there was a greater reduction in infarct size (-32.4 ± 33 vs. -12.8 ± 24%; p = 0.003) and a significant improvement in left ventricular (LV) volumes and ejection fraction. Level of circulating CD34+/VEGF-R2+ cells was correlated with the number of ECFC colonies (r = 0.54, p < 0.001) and relative change in infarct size (r = 0.71, p < 0.0001). The results showed that the presence of ECFC colonies is associated with reduced MVO after AMI, leading to reduced infarct size and less LV remodelling and can be considered a marker of preserved microvascular integrity in AMI patients.
Collapse
|
32
|
Ahrens I, Domeij H, Eisenhardt SU, Topcic D, Albrecht M, Leitner E, Viitaniemi K, Jowett JB, Lappas M, Bode C, Haviv I, Peter K. Opposing effects of monomeric and pentameric C-reactive protein on endothelial progenitor cells. Basic Res Cardiol 2011; 106:879-95. [PMID: 21562922 PMCID: PMC3149664 DOI: 10.1007/s00395-011-0191-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 04/12/2011] [Accepted: 04/29/2011] [Indexed: 12/17/2022]
Abstract
C-reactive protein (CRP) has been linked to the pathogenesis of atherosclerosis. The dissociation of native, pentameric (p)CRP to monomeric (m)CRP on the cell membrane of activated platelets has recently been demonstrated. The dissociation of pCRP to mCRP may explain local pro-inflammatory reactions at the site of developing atherosclerotic plaques. As a biomarker, pCRP predicts cardiovascular adverse events and so do reduced levels and function of circulating endothelial progenitor cells (EPCs). We hypothesised that mCRP and pCRP exert a differential effect on EPC function and differentiation. EPCs were treated with mCRP or pCRP for 72 h, respectively. Phenotypical characterisation was done by flow cytometry and immunofluorescence microscopy, while the effect of mCRP and pCRP on gene expression was examined by whole-genome gene expression analysis. The functional capacity of EPCs was determined by colony forming unit (CFU) assay and endothelial tube formation assay. Double staining for acetylated LDL and ulex lectin significantly decreased in cells treated with pCRP. The length of tubuli in a matrigel assay with HUVECs decreased significantly in response to pCRP, but not to mCRP. The number of CFUs increased after pCRP treatment. RNA expression profiling demonstrated that mCRP and pCRP cause highly contradictory gene regulation. Interferon-responsive genes (IFI44L, IFI44, IFI27, IFI 6, MX1, OAS2) were among the highly up-regulated genes after mCRP, but not after pCRP treatment. In conclusion, EPC phenotype, genotype and function were differentially affected by mCRP and pCRP, strongly arguing for differential roles of these two CRP conformations. The up-regulation of interferon-inducible genes in response to mCRP may constitute a mechanism for the local regulation of EPC function.
Collapse
Affiliation(s)
- I Ahrens
- Department of Cardiology and Angiology, University Hospital Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lau LF. CCN1/CYR61: the very model of a modern matricellular protein. Cell Mol Life Sci 2011; 68:3149-63. [PMID: 21805345 DOI: 10.1007/s00018-011-0778-3] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 07/19/2011] [Accepted: 07/19/2011] [Indexed: 02/08/2023]
Abstract
CCN1 (CYR61) is a dynamically expressed, multifunctional matricellular protein that plays essential roles in cardiovascular development during embryogenesis, and regulates inflammation, wound healing and fibrogenesis in the adult. Aberrant CCN1 expression is associated with myriad pathologies, including various cancers and diseases associated with chronic inflammation. CCN1 promotes diverse and sometimes opposing cellular responses, which can be ascribed, as least in part, to disparate activities mediated through its direct binding to distinct integrins in different cell types and contexts. Accordingly, CCN1 promotes cell proliferation, survival and angiogenesis by binding to integrin α(v)β(3), and induces apoptosis and senescence through integrin α(6)β(1) and heparan sulfate proteoglycans. The ability of CCN1 to trigger the accumulation of a robust and sustained level of reactive oxygen species underlies some of its unique activities as a matrix cell-adhesion molecule. Emerging studies suggest that CCN1 might be useful as a biomarker or therapeutic target in certain diseases.
Collapse
Affiliation(s)
- Lester F Lau
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, 900 S. Ashland Avenue, Chicago, IL 60607, USA.
| |
Collapse
|
34
|
Therapeutic neovascularization for coronary disease: current state and future prospects. Basic Res Cardiol 2011; 106:897-909. [DOI: 10.1007/s00395-011-0200-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 06/01/2011] [Accepted: 06/17/2011] [Indexed: 12/19/2022]
|