1
|
Shen S, Pan T, Liu P, Tian Y, Shi Y, Zhu W. The mechanisms and applications of endothelial progenitor cell therapy in the treatment of intracranial aneurysm. J Transl Med 2025; 23:377. [PMID: 40148864 PMCID: PMC11951544 DOI: 10.1186/s12967-025-06401-w] [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] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The pathophysiological mechanism of intracranial aneurysm (IA) involves the dynamic interaction of ECM abnormalities, hemodynamic stress, and inflammatory response. The rupture of intracranial aneurysm will cause serious consequences. Multiple studies have confirmed the important role and potential application of endothelial progenitor cells (EPCs) in vascular repair. This review focuses on the specific mechanism of EPCs in the treatment of intracranial aneurysms, which promote re-endothelialization and angiogenesis through bone marrow mobilization, targeted migration to the site of injury, differentiation into mature endothelial cells, and secretion of angiogenic factors. In addition, EPCs maintain ECM homeostasis by regulating MMP/IMP balance, inhibiting aneurysm wall thinning and structural damage. Based on the vascular repair mechanism of EPCs, new treatment strategies such as "biologically active" spring coils (loaded with EPCs or SDF-1α) and flow diverters(FDs) combined with EPCs therapy have been developed to synergistically promote carotid endothelialization of aneurysms and reduce the risk of recurrence. Future research needs to further validate the long-term efficacy and precise regulatory mechanisms of EPCs in clinical translation, providing new directions for IA treatment.
Collapse
Affiliation(s)
- Shiyu Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Tonglin Pan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yanlong Tian
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yuan Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
2
|
Yu J, Du Q, Li X, Wei W, Fan Y, Zhang J, Chen J. Potential role of endothelial progenitor cells in the pathogenesis and treatment of cerebral aneurysm. Front Cell Neurosci 2024; 18:1456775. [PMID: 39193428 PMCID: PMC11348393 DOI: 10.3389/fncel.2024.1456775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Cerebral aneurysm (CA) is a significant health concern that results from pathological dilations of blood vessels in the brain and can lead to severe and potentially life-threatening conditions. While the pathogenesis of CA is complex, emerging studies suggest that endothelial progenitor cells (EPCs) play a crucial role. In this paper, we conducted a comprehensive literature review to investigate the potential role of EPCs in the pathogenesis and treatment of CA. Current research indicates that a decreased count and dysfunction of EPCs disrupt the balance between endothelial dysfunction and repair, thus increasing the risk of CA formation. Reversing these EPCs abnormalities may reduce the progression of vascular degeneration after aneurysm induction, indicating EPCs as a promising target for developing new therapeutic strategies to facilitate CA repair. This has motivated researchers to develop novel treatment options, including drug applications, endovascular-combined and tissue engineering therapies. Although preclinical studies have shown promising results, there is still a considerable way to go before clinical translation and eventual benefits for patients. Nonetheless, these findings offer hope for improving the treatment and management of this condition.
Collapse
Affiliation(s)
- Jin Yu
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Du
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuncun Fan
- Department of Respiratory and Critical Care Medicine, Laifeng County People’s Hospital, Enshi, Hubei, China
| | - Jianjian Zhang
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jincao Chen
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
3
|
Tang Y, Yin L, Gao S, Long X, Du Z, Zhou Y, Zhao S, Cao Y, Pan S. A small-diameter vascular graft immobilized peptides for capturing endothelial colony-forming cells. Front Bioeng Biotechnol 2023; 11:1154986. [PMID: 37101749 PMCID: PMC10123284 DOI: 10.3389/fbioe.2023.1154986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 04/28/2023] Open
Abstract
Combining synthetic polymers and biomacromolecules prevents the occurrence of thrombogenicity and intimal hyperplasia in small-diameter vascular grafts (SDVGs). In the present study, an electrospinning poly (L)-lactic acid (PLLA) bilayered scaffold is developed to prevent thrombosis after implantation by promoting the capture and differentiation of endothelial colony-forming cells (ECFCs). The scaffold consists of an outer PLLA scaffold and an inner porous PLLA biomimetic membrane combined with heparin (Hep), peptide Gly-Gly-Gly-Arg-Glu-Asp-Val (GGG-REDV), and vascular endothelial growth factor (VEGF). Attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy, X-ray photoelectron spectroscopy (XPS), and contact angle goniometry were performed to determine successful synthesis. The tensile strength of the outer layer was obtained using the recorded stress/strain curves, and hemocompatibility was evaluated using the blood clotting test. The proliferation, function, and differentiation properties of ECFCs were measured on various surfaces. Scanning electronic microscopy (SEM) was used to observe the morphology of ECFCs on the surface. The outer layer of scaffolds exhibited a similar strain and stress performance as the human saphenous vein via the tensile experiment. The contact angle decreased continuously until it reached 56° after REDV/VEGF modification, and SEM images of platelet adhesion showed a better hemocompatibility surface after modification. The ECFCs were captured using the REDV + VEGF + surface successfully under flow conditions. The expression of mature ECs was constantly increased with the culture of ECFCs on REDV + VEGF + surfaces. SEM images showed that the ECFCs captured by the REDV + VEGF + surface formed capillary-like structures after 4 weeks of culture. The SDVGs modified by REDV combined with VEGF promoted ECFC capture and rapid differentiation into ECs, forming capillary-like structures in vitro. The bilayered SDVGs could be used as vascular devices that achieved a high patency rate and rapid re-endothelialization.
Collapse
Affiliation(s)
- Yaqi Tang
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Lu Yin
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, China
| | - Shuai Gao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Xiaojing Long
- State Key Laboratory of Bio-fibers and Eco-textiles, Qingdao University, Qingdao, China
| | - Zhanhui Du
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Yingchao Zhou
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Shuiyan Zhao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Yue Cao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Silin Pan
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Baldassarro VA, Giraldi V, Giuliani A, Moretti M, Pagnotta G, Flagelli A, Clavenzani P, Lorenzini L, Giardino L, Focarete ML, Giacomini D, Calzà L. Poly(l-lactic acid) Scaffold Releasing an α 4β 1 Integrin Agonist Promotes Nonfibrotic Skin Wound Healing in Diabetic Mice. ACS APPLIED BIO MATERIALS 2022; 6:296-308. [PMID: 36542733 PMCID: PMC9937562 DOI: 10.1021/acsabm.2c00890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Skin wound healing is a highly complex process that continues to represent a major medical problem, due to chronic nonhealing wounds in several classes of patients and to possible fibrotic complications, which compromise the function of the dermis. Integrins are transmembrane receptors that play key roles in this process and that offer a recognized druggable target. Our group recently synthesized GM18, a specific agonist for α4β1, an integrin that plays a role in skin immunity and in the migration of neutrophils, also regulating the differentiated state of fibroblasts. GM18 can be combined with poly(l-lactic acid) (PLLA) nanofibers to provide a controlled release of this agonist, resulting in a medication particularly suitable for skin wounds. In this study, we first optimized a GM18-PLLA nanofiber combination with a 7-day sustained release for use as skin wound medication. When tested in an experimental pressure ulcer in diabetic mice, a model for chronic nonhealing wounds, both soluble and GM18-PLLA formulations accelerated wound healing, as well as regulated extracellular matrix synthesis toward a nonfibrotic molecular signature. In vitro experiments using the adhesion test showed fibroblasts to be a principal GM18 cellular target, which we then used as an in vitro model to explore possible mechanisms of GM18 action. Our results suggest that the observed antifibrotic behavior of GM18 may exert a dual action on fibroblasts at the α4β1 binding site and that GM18 may prevent profibrotic EDA-fibronectin-α4β1 binding and activate outside-in signaling of the ERK1/2 pathways, a critical component of the wound healing process.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Valentina Giraldi
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Alessandro Giuliani
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Marzia Moretti
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Giorgia Pagnotta
- Department
of Chemistry “Giacomo Ciamician” and INSTM UdR of Bologna, University of Bologna, 2 via Selmi, 40126 Bologna, Italy
| | - Alessandra Flagelli
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Paolo Clavenzani
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Luca Lorenzini
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Luciana Giardino
- Department
of Veterinary Medical Science, University
of Bologna, 50 Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,IRET
Foundation, 41/E Via
Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy
| | - Maria Letizia Focarete
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Department
of Chemistry “Giacomo Ciamician” and INSTM UdR of Bologna, University of Bologna, 2 via Selmi, 40126 Bologna, Italy
| | - Daria Giacomini
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Department
of Chemistry “Giacomo Ciamician” and INSTM UdR of Bologna, University of Bologna, 2 via Selmi, 40126 Bologna, Italy,
| | - Laura Calzà
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, 41/E Via Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,IRET
Foundation, 41/E Via
Tolara di Sopra, 40064 Ozzano Emilia, Bologna, Italy,Department
of Pharmacy and BioTechnology, University
of Bologna, 15 Via San
Donato, 40127 Bologna, Italy,
| |
Collapse
|
5
|
Vithani V, Sutariya B, Montenegro DM, Chukwu M, Ehsan P, Aburumman RN, Muthanna SI, Menon SR, Penumetcha SS. A Systematic Review of CD34+ Stem Cell Therapy as an Innovative and Efficient Treatment for the Management of Refractory Angina. Cureus 2022; 14:e32665. [PMID: 36660500 PMCID: PMC9844930 DOI: 10.7759/cureus.32665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/18/2022] [Indexed: 12/23/2022] Open
Abstract
Despite optimal medical treatment, many individuals suffering from severe coronary artery disease are not suitable candidates for further revascularization. Therapeutic angiogenesis has attracted continuous interest to increase myocardial perfusion. Cell therapy using autologous stem cells expressing Cluster of Differentiation 34 plus (CD34+) offers a special therapeutic choice for individuals with refractory angina, seeing as CD34+ stem cells can restore microcirculation. We searched PubMed, PubMed Central (PMC), and Google Scholar to find the relevant articles to write this systematic review about the role of CD34+ stem cell therapy in the management of refractory angina. Additionally, we provided a brief explanation of CD34+ cells and their mechanism of action. Along with the positive finding of other trials, a recent open-label, single-center intracoronary CD34+ cell therapy for the treatment of coronary endothelial dysfunction in patients with angina and nonobstructive coronary arteries (IMPROvE-CED) clinical trial published in 2022 concluded improvement in coronary blood flow, a significant reduction in daily as-needed sublingual nitroglycerin use and improvement in Canadian Cardiovascular Society (CCS) angina class were observed after autologous CD34+ cell treatment. In conclusion, refractory angina management and overall prognosis may be revolutionized once this treatment is approved.
Collapse
Affiliation(s)
- Vruti Vithani
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Medicine, Government Medical College, Surat, IND
| | - Bansi Sutariya
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Medicine, Government Medical College, Surat, IND
| | - Diana M Montenegro
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Michael Chukwu
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Surgery, Pilgrim Hospital, Boston, GBR
| | - Paghunda Ehsan
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Hayatabad Medical Complex Peshawar, Peshawar, PAK
| | - Rawia N Aburumman
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Mu'tah University, Amman, JOR
| | - Shivani Ishwarya Muthanna
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bengaluru, IND
| | | | - Sai Sri Penumetcha
- General Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Medicine, Chalmeda Anand Rao Institute of Medical Sciences, Karimnagar, IND
| |
Collapse
|
6
|
Cao L, Dong Y, Sun K, Li D, Wang H, Li H, Yang B. Experimental Animal Models for Moyamoya Disease: A Species-Oriented Scoping Review. Front Surg 2022; 9:929871. [PMID: 35846951 PMCID: PMC9283787 DOI: 10.3389/fsurg.2022.929871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Moyamoya disease (MMD) is a rare cerebrovascular disease characterized by progressive stenosis of large intracranial arteries and a hazy network of basal collaterals called moyamoya vessels. The etiology and pathogenesis of MMD are still obscure. The biggest obstacles in the basic research of MMD are difficulty in obtaining specimens and the lack of an animal model. It is necessary to use appropriate and rationally designed animal models for the correct evaluation. Several animal models and methods have been developed to produce an effective MMD model, such as zebrafish, mice and rats, rabbits, primates, felines, canines, and peripheral blood cells, each with advantages and disadvantages. There are three mechanisms for developing animal models, including genetic, immunological/inflammatory, and ischemic animal models. This review aims to analyze the characteristics of currently available models, providing an overview of the animal models framework and the convenience of selecting model types for MMD research. It will be a great benefit to identify strategies for future model generations.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Zhu J, Tan J, Zhang C, Jia J, Leng H, Xu Y, Song C. Single Intraosseous Simvastatin Application Induces Endothelial Progenitor Cell Mobilization and Therapeutic Angiogenesis in a Diabetic Hindlimb Ischemia Rat Model. Plast Reconstr Surg 2021; 148:936e-945e. [PMID: 34644264 DOI: 10.1097/prs.0000000000008526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Endothelial progenitor cells have shown the ability to enhance neovascularization. In this study, the authors tested whether intraosseous delivery of simvastatin could mobilize endothelial progenitor cells and enhance recovery in a hindlimb ischemia model. METHODS There are eight groups of rats in this study: normal control; type 1 diabetes mellitus control group control without drug intervention; and type 1 diabetes mellitus rats that randomly received intraosseous simvastatin (0, 0.5, or 1 mg) or oral simvastatin administration (0, 20, or 400 mg). All type 1 diabetes mellitus rats had induced hindlimb ischemia. The number of endothelial progenitor cells in peripheral blood, and serum markers, were detected. The recovery of blood flow at 21 days after treatment was used as the main outcome. RESULTS The authors demonstrated that endothelial progenitor cell mobilization was increased in the simvastatin 0.5- and 1-mg groups compared with the type 1 diabetes mellitus control and simvastatin 0-mg groups at 1, 2, and 3 weeks. Serum vascular endothelial growth factor levels were significantly increased at 2 weeks in the simvastatin 0.5- and 1-mg groups, in addition to the increase of the blood flow and the gastrocnemius weight at 3 weeks. Similar increase can also been seen in simvastatin 400 mg orally but not in simvastatin 20 mg orally. CONCLUSION These findings demonstrate that a single intraosseous administration of simvastatin mobilized endothelial progenitor cells at a dose one-hundredth of the required daily oral dose in rats, and this potent mobilization of endothelial progenitor cells markedly improved diabetic limb ischemia by means of neovascularization.
Collapse
Affiliation(s)
- Junxiong Zhu
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Jie Tan
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Chenggui Zhang
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Jialin Jia
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Huijie Leng
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Yingsheng Xu
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - ChunLi Song
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| |
Collapse
|
8
|
Hao J, Zhou H, Nemes K, Yen D, Zhao W, Bramlett C, Wang B, Lu R, Shen K. Membrane-bound SCF and VCAM-1 synergistically regulate the morphology of hematopoietic stem cells. J Cell Biol 2021; 220:212562. [PMID: 34402812 PMCID: PMC8374872 DOI: 10.1083/jcb.202010118] [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: 10/20/2020] [Revised: 06/29/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Membrane-bound factors expressed by niche stromal cells constitute a unique class of localized cues and regulate the long-term functions of adult stem cells, yet little is known about the underlying mechanisms. Here, we used a supported lipid bilayer (SLB) to recapitulate the membrane-bound interactions between hematopoietic stem cells (HSCs) and niche stromal cells. HSCs cluster membrane-bound stem cell factor (mSCF) at the HSC-SLB interface. They further form a polarized morphology with aggregated mSCF under a large protrusion through a synergy with VCAM-1 on the bilayer, which drastically enhances HSC adhesion. These features are unique to mSCF and HSCs among the factors and hematopoietic populations we examined. The mSCF-VCAM-1 synergy and the polarized HSC morphology require PI3K signaling and cytoskeletal reorganization. The synergy also enhances nuclear retention of FOXO3a, a crucial factor for HSC maintenance, and minimizes its loss induced by soluble SCF. Our work thus reveals a unique role and signaling mechanism of membrane-bound factors in regulating stem cell morphology and function.
Collapse
Affiliation(s)
- Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Kristen Nemes
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Daniel Yen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Winfield Zhao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Charles Bramlett
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - Bowen Wang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA.,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.,Department of Medicine, University of Southern California, Los Angeles, CA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.,USC Stem Cell, University of Southern California, Los Angeles, CA
| |
Collapse
|
9
|
Role of Stromal Cell-Derived Factor-1 in Endothelial Progenitor Cell-Mediated Vascular Repair and Regeneration. Tissue Eng Regen Med 2021; 18:747-758. [PMID: 34449064 PMCID: PMC8440704 DOI: 10.1007/s13770-021-00366-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are immature endothelial cells that participate in vascular repair and postnatal neovascularization and provide a novel and promising therapy for the treatment of vascular disease. Studies in different animal models have shown that EPC mobilization through pharmacological agents and autologous EPC transplantation contribute to restoring blood supply and tissue regeneration after ischemic injury. However, these effects of the progenitor cells in clinical studies exhibit mixed results. The therapeutic efficacy of EPCs is closely associated with the number of the progenitor cells recruited into ischemic regions and their functional abilities and survival in injury tissues. In this review, we discussed the regulating role of stromal cell-derived factor-1 (also known CXCL12, SDF-1) in EPC mobilization, recruitment, homing, vascular repair and neovascularization, and analyzed the underlying machemisms of these functions. Application of SDF-1 to improve the regenerative function of EPCs following vascular injury was also discussed. SDF-1 plays a crucial role in mobilizing EPC from bone marrow into peripheral circulation, recruiting the progenitor cells to target tissue and protecting against cell death under pathological conditions; thus improve EPC regenerative capacity. SDF-1 are crucial for regulating EPC regenerative function, and provide a potential target for improve therapeutic efficacy of the progenitor cells in treatment of vascular disease.
Collapse
|
10
|
Neovascularization and tissue regeneration by endothelial progenitor cells in ischemic stroke. Neurol Sci 2021; 42:3585-3593. [PMID: 34216308 DOI: 10.1007/s10072-021-05428-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/19/2021] [Indexed: 12/26/2022]
Abstract
Endothelial progenitor cells (EPCs) are immature endothelial cells (ECs) capable of proliferating and differentiating into mature ECs. These progenitor cells migrate from bone marrow (BM) after vascular injury to ischemic areas, where they participate in the repair of injured endothelium and new blood vessel formation. EPCs also secrete a series of protective cytokines and growth factors that support cell survival and tissue regeneration. Thus, EPCs provide novel and promising potential therapies to treat vascular disease, including ischemic stroke. However, EPCs are tightly regulated during the process of vascular repair and regeneration by numerous endogenous cytokines that are associated closely with the therapeutic efficacy of the progenitor cells. The regenerative capacity of EPCs also is affected by a range of exogenous factors and drugs as well as vascular risk factors. Understanding the functional properties of EPCs and the factors related to their regenerative capacity will facilitate better use of these progenitor cells in treating vascular disease. Here, we review the current knowledge of EPCs in cerebral neovascularization and tissue regeneration after cerebral ischemia and the factors associated with their regenerative function to better understand the underlying mechanisms and provide more effective strategies for the use of EPCs in treating ischemic stroke.
Collapse
|
11
|
Yu J, Du Q, Hu M, Zhang J, Chen J. Endothelial Progenitor Cells in Moyamoya Disease: Current Situation and Controversial Issues. Cell Transplant 2021; 29:963689720913259. [PMID: 32193953 PMCID: PMC7444216 DOI: 10.1177/0963689720913259] [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] [Indexed: 12/12/2022] Open
Abstract
Due to the lack of animal models and difficulty in obtaining specimens, the study of pathogenesis of moyamoya disease (MMD) almost stagnated. In recent years, endothelial progenitor cells (EPCs) have attracted more and more attention in vascular diseases due to their important role in neovascularization. With the aid of paradigms and methods in cardiovascular diseases research, people began to explore the role of EPCs in the processing of MMD. In the past decade, studies have shown that abnormalities in cell amounts and functions of EPCs were closely related to the vascular pathological changes in MMD. However, the lack of consistent criteria, such as isolation, cultivation, and identification standards, is also blocking the way forward. The goal of this review is to provide an overview of the current situation and controversial issues relevant to studies about EPCs in the pathogenesis and etiology of MMD.
Collapse
Affiliation(s)
- Jin Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qian Du
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Miao Hu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianjian Zhang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jincao Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Matta A, Nader V, Galinier M, Roncalli J. Transplantation of CD34+ cells for myocardial ischemia. World J Transplant 2021; 11:138-146. [PMID: 34046316 PMCID: PMC8131931 DOI: 10.5500/wjt.v11.i5.138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
CD34+ cells are multipotent hematopoietic stem cells also known as endothelial progenitor cells and are useful in regenerative medicine. Naturally, these cells are mobilized from the bone marrow into peripheral circulation in response to ischemic tissue injury. CD34+ cells are known for their high proliferative and differentiation capacities that play a crucial role in the repair process of myocardial damage. They have an important paracrine activity in secreting factors to stimulate vasculogenesis, reduce endothelial cells and cardiomyocytes apoptosis, remodel extracellular matrix and activate additional progenitor cells. Once they migrate to the target site, they enhance angiogenesis, neovascularization and tissue regeneration. Several trials have demonstrated the safety and efficacy of CD34+ cell therapy in different settings, such as peripheral limb ischemia, stroke and cardiovascular disease. Herein, we review the potential utility of CD34+ cell transplantation in acute myocardial infarction, refractory angina and ischemic heart failure.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, Toulouse 31059, France
- Faculty of Medicine, Holy Spirit University of Kaslik, Kaslik 00000, Lebanon
| | - Vanessa Nader
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, Toulouse 31059, France
- Faculty of Pharmacy, Lebanese University, Beirut 961, Lebanon
| | - Michel Galinier
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, Toulouse 31059, France
| | - Jerome Roncalli
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, Toulouse 31059, France
| |
Collapse
|
13
|
Zhang E, Liu Y, Han C, Fan C, Wang L, Chen W, Du Y, Han D, Arnone B, Xu S, Wei Y, Mobley J, Qin G. Visualization and Identification of Bioorthogonally Labeled Exosome Proteins Following Systemic Administration in Mice. Front Cell Dev Biol 2021; 9:657456. [PMID: 33898459 PMCID: PMC8058422 DOI: 10.3389/fcell.2021.657456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Exosomes transport biologically active cargo (e.g., proteins and microRNA) between cells, including many of the paracrine factors that mediate the beneficial effects associated with stem-cell therapy. Stem cell derived exosomes, in particular mesenchymal stem cells (MSCs), have been shown previously to largely replicate the therapeutic activity associated with the cells themselves, which suggests that exosomes may be a useful cell-free alternative for the treatment of cardiovascular disorders. However, the mechanisms that govern how exosomes home to damaged cells and tissues or the uptake and distribution of exosomal cargo are poorly characterized, because techniques for distinguishing between exosomal proteins and proteins in the targeted tissues are lacking. Here, we report the development of an in vivo model that enabled the visualization, tracking, and quantification of proteins from systemically administered MSC exosomes. The model uses bioorthogonal chemistry and cell-selective metabolic labeling to incorporate the non-canonical amino acid azidonorleucine (ANL) into the MSC proteome. ANL incorporation is facilitated via expression of a mutant (L274G) methionyl-tRNA-synthetase (MetRS∗) and subsequent incubation with ANL-supplemented media; after which ANL can be covalently linked to alkyne-conjugated reagents (e.g., dyes and resins) via click chemistry. Our results demonstrate that when the exosomes produced by ANL-treated, MetRS∗-expressing MSCs were systemically administered to mice, the ANL-labeled exosomal proteins could be accurately and reliably identified, isolated, and quantified from a variety of mouse organs, and that myocardial infarction (MI) both increased the abundance of exosomal proteins and redistributed a number of them from the membrane fraction of intact hearts to the cytosol of cells in infarcted hearts. Additionally, we found that Desmoglein-1c is enriched in MSC exosomes and taken up by ischemic myocardium. Collectively, our results indicate that this newly developed bioorthogonal system can provide crucial insights into exosome homing, as well as the uptake and biodistribution of exosomal proteins.
Collapse
Affiliation(s)
- Eric Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yanwen Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chaoshan Han
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chengming Fan
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lu Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Wangping Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yipeng Du
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Dunzheng Han
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Baron Arnone
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shiyue Xu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James Mobley
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, School of Medicine, Birmingham, AL, United States
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
14
|
Han D, Yang J, Zhang E, Liu Y, Boriboun C, Qiao A, Yu Y, Sun J, Xu S, Yang L, Yan W, Luo B, Lu D, Zhang C, Jie C, Mobley J, Zhang J, Qin G. Analysis of mesenchymal stem cell proteomes in situ in the ischemic heart. Am J Cancer Res 2020; 10:11324-11338. [PMID: 33042285 PMCID: PMC7532665 DOI: 10.7150/thno.47893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Cell therapy for myocardial infarction is promising but largely unsuccessful in part due to a lack of mechanistic understanding. Techniques enabling identification of stem cell-specific proteomes in situ in the injured heart may shed light on how the administered cells respond to the injured microenvironment and exert reparative effects. Objective: To identify the proteomes of the transplanted mesenchymal stem cells (MSCs) in the infarcted myocardium, we sought to target a mutant methionyl-tRNA synthetase (MetRSL274G) in MSCs, which charges azidonorleucine (ANL), a methionine analogue and non-canonical amino acid, to tRNA and subsequently to nascent proteins, permitting isolation of ANL-labeled MSC proteomes from ischemic hearts by ANL-alkyne based click reaction. Methods and Results: Murine MSCs were transduced with lentivirus MetRSL274G and supplemented with ANL; the ANL-tagged nascent proteins were visualized by bio-orthogonal non-canonical amino-acid tagging, spanning all molecular weights and by fluorescent non-canonical amino-acid tagging, displaying strong fluorescent signal. Then, the MetRSL274G-transduced MSCs were administered to the infarcted or Sham heart in mice receiving ANL treatment. The MSC proteomes were isolated from the left ventricular protein lysates by click reaction at days 1, 3, and 7 after cell administration, identified by LC/MS. Among all identified proteins (in Sham and MI hearts, three time-points each), 648 were shared by all 6 groups, accounting for 82±5% of total proteins in each group, and enriched under mitochondrion, extracellular exosomes, oxidation-reduction process and poly(A) RNA binding. Notably, 26, 110 and 65 proteins were significantly up-regulated and 11, 28 and 19 proteins were down-regulated in the infarcted vs. Sham heart at the three time-points, respectively; these proteins are pronounced in the GO terms of extracellular matrix organization, response to stress and regulation of apoptotic process and in the KEGG pathways of complements and coagulation cascades, apoptosis, and regulators of actin cytoskeleton. Conclusions: MetRSL274G expression allows successful identification of MSC-specific nascent proteins in the infarcted hearts, which reflect the functional states, adaptive response, and reparative effects of MSCs that may be leveraged to improve cardiac repair.
Collapse
|
15
|
Zheng J, Chen M, Ye C, Sun X, Jiang N, Zou X, Yang H, Liu H. BuZangTongLuo decoction improved hindlimb ischemia by activating angiogenesis and regulating gut microbiota in diabetic mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112330. [PMID: 31654796 DOI: 10.1016/j.jep.2019.112330] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/20/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Seven traditional medicinal plants (including Astragalus membranaceus, Dioscorea hemsleyi, Salvia miltiorrhiza, Scrophularia ningpoensis, Ophiopogon japonicus, Panax ginseng and Fritillariae cirrhosae) and one insect leech (Whitmania pigra Whitman) were combined into BuZangTongLuo formula (BZTLF) under the guidance of traditional Chinese medicine. BZTLF is potentially effective against diabetic vascular complications. AIM OF THE STUDY Previous studies failed to clarify the molecular mechanism through which BZTLF suppressed diabetic ischemia. In this study, we aimed to explore whether BZTLF treatment could prevent the occurrence of type 2 diabetic (T2D) hindlimb ischemia in mice. Further, we investigated the regulatory effect of BZTLF on angiogenesis-related VEGF signaling pathway and gut microbiota dysfunction in diabetic ischemia mice. MATERIALS AND METHODS C57BL/6J mice fed with high-fat diet (HFD) received STZ injection and femoral artery ligation to build T2D diabetic hindlimb ischemia model. Mice were gavaged with BZTLF (5 g [raw materials]/kg/d) or with metformin plus atorvastatin for three weeks. Laser doppler imaging system was utilized for the visualization of blood flow. Histochemistry analysis was performed for microvascular vessel staining. Western blot was applied to detect the protein changes of signaling molecules responsible for VEGF pathway. Finally, 16S rDNA gene sequencing was conducted for analysis of gut microbiota structure. RESULTS BZTLF treatment remarkably restored blood flow and capillary density of diabetic hindlimb ischemia. And the protein changes of VEGF signaling molecules were reversed in BZTLF-treated diabetic ischemia mice, including the decreased VEGF and HIF-1α, and the increased NO, eNOS and p-ERK1/2. The gut microbiota analysis suggests that BZTLF treatment increased the abundances of several beneficial bacteria (Akkermansia, Bifidobacterium and Bacteroides), while decreased the populations of some harmful bacteria(Blautia, Weissella, Escherichia Shigella and Kurthia). By using Spearman's correlation analysis, these changed gut flora were positively/negatively correlated with VEGF signaling pathway or glycometabolic parameters. CONCLUSION BZTLF displayed beneficial effects on diabetic hindlimb ischemia by reshaping the gut microbiota structure and stunning the VEGF/HIF-1α pathway.
Collapse
MESH Headings
- Animals
- Blood Flow Velocity
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/microbiology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Angiopathies/drug therapy
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/microbiology
- Diabetic Angiopathies/physiopathology
- Drugs, Chinese Herbal/pharmacology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Gastrointestinal Microbiome/drug effects
- Hindlimb/blood supply
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Ischemia/drug therapy
- Ischemia/metabolism
- Ischemia/microbiology
- Ischemia/physiopathology
- Male
- Mice, Inbred C57BL
- Neovascularization, Physiologic/drug effects
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Regional Blood Flow
- Signal Transduction
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Junping Zheng
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China; College of Life Sciences, Wuchang University of Technology, Wuhan, 430223, China
| | - Man Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Cheng Ye
- Wuhan Customs Technology Center, Wuhan, 430050, China
| | - Xiongjie Sun
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Nan Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Xiaojuan Zou
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Huabing Yang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Hongtao Liu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China; College of Life Sciences, Wuchang University of Technology, Wuhan, 430223, China.
| |
Collapse
|
16
|
Filippi A, Constantin A, Alexandru N, Voicu G, Constantinescu CA, Rebleanu D, Fenyo M, Simionescu D, Simionescu A, Manduteanu I, Georgescu A. Integrins α4β1 and αVβ3 are Reduced in Endothelial Progenitor Cells from Diabetic Dyslipidemic Mice and May Represent New Targets for Therapy in Aortic Valve Disease. Cell Transplant 2020; 29:963689720946277. [PMID: 32841051 PMCID: PMC7563030 DOI: 10.1177/0963689720946277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes reduces the number and induces dysfunction in circulating endothelial progenitor cells (EPCs) by mechanisms that are still uncovered. This study aims to evaluate the number, viability, phenotype, and function of EPCs in dyslipidemic mice with early diabetes mellitus and EPC infiltration in the aortic valve in order to identify possible therapeutic targets in diabetes-associated cardiovascular disease. A streptozotocin-induced diabetic apolipoprotein E knock-out (ApoE-/-) mouse model was used to identify the early and progressive changes, at 4 or 7 days on atherogenic diet after the last streptozotocin or citrate buffer injection. Blood and aortic valves from diabetic or nondiabetic ApoE-/- animals were collected.EPCs were identified as CD34 and vascular endothelial growth factor receptor 2 positive monocytes, and the expression levels of α4β1, αVβ3, αVβ5, β1, αLβ2, α5 integrins, and C-X-C chemokine receptor type 4 chemokine receptor on EPC surface were assessed by flow cytometry. The number of CD34 positive cells in the aortic valve, previously found to be recruited progenitor cells, was measured by fluorescence microscopy. Our results show that aortic valves from mice fed 7 days with atherogenic diet presented a significantly higher number of CD34 positive cells compared with mice fed only 4 days with the same diet, and diabetes reversed this finding. We also show a reduction of circulatory EPC numbers in diabetic mice caused by cell senescence and lower mobilization. Dyslipidemia induced EPC death through apoptosis regardless of the presence of diabetes, as shown by the higher percent of propidium iodide positive cells and higher cleaved caspase-3 levels. EPCs from diabetic mice expressed α4β1 and αVβ3 integrins at a lower level, while the rest of the integrins tested were unaffected by diabetes or diet. In conclusion, reduced EPC number and expression of α4β1 and αVβ3 integrins on EPCs at 4 and 7 days after diabetes induction in atherosclerosis-prone mice have resulted in lower recruitment of EPCs in the aortic valve.
Collapse
Affiliation(s)
- Alexandru Filippi
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | - Alina Constantin
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | - Nicoleta Alexandru
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | - Geanina Voicu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | | | - Daniela Rebleanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | - Madalina Fenyo
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | | | - Agneta Simionescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
- Clemson University, Clemson SC, USA
| | - Ileana Manduteanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| | - Adriana Georgescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of
Romanian Academy, Bucharest, Romania
| |
Collapse
|
17
|
Han S, Xu S, Zhou J, Qiao A, Boriboun C, Ma W, Li H, Biyashev D, Yang L, Zhang E, Liu Q, Jiang S, Zhao TC, Krishnamurthy P, Zhang C, Richard S, Qiu H, Zhang J, Qin G. Sam68 impedes the recovery of arterial injury by augmenting inflammatory response. J Mol Cell Cardiol 2019; 137:82-92. [PMID: 31639388 DOI: 10.1016/j.yjmcc.2019.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/23/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The role of Src-associated-in-mitosis-68-kDa (Sam68) in cardiovascular biology has not been studied. A recent report suggests that Sam68 promotes TNF-α-induced NF-κB activation in fibroblasts. Here we sought to dissect the molecular mechanism by which Sam68 regulates NF-κB signaling and its functional significance in vascular injury. APPROACH AND RESULTS The endothelial denudation injury was induced in the carotid artery of Sam68-null (Sam68-/-) and WT mice. Sam68-/- mice displayed an accelerated re-endothelialization and attenuated neointima hyperplasia, which was associated with a reduced macrophage infiltration and lowered expression of pro-inflammatory cytokines in the injured vessels. Remarkably, the ameliorated vascular remodeling was recapitulated in WT mice after receiving transplantation of bone marrow (BM) from Sam68-/- mice, suggesting the effect was attributable to BM-derived inflammatory cells. In cultured Raw264.7 macrophages, knockdown of Sam68 resulted in a significant reduction in the TNF-α-induced expression of TNF-α, IL-1β, and IL-6 and in the level of nuclear phospho-p65, indicating attenuated NF-κB activation; and these results were confirmed in peritoneal and BM-derived macrophages of Sam68-/- vs. WT mice. Furthermore, co-immunoprecipitation and mass-spectrometry identified Filamin A (FLNA) as a novel Sam68-interacting protein upon TNF-α treatment. Loss- and gain-of-function experiments suggest that Sam68 and FLNA are mutually dependent for NF-κB activation and pro-inflammatory cytokine expression, and that the N-terminus of Sam68 is required for TRAF2-FLNA interaction. CONCLUSIONS Sam68 promotes pro-inflammatory response in injured arteries and impedes recovery by interacting with FLNA to stabilize TRAF2 on the cytoskeleton and consequently potentiate NF-κB signaling.
Collapse
Affiliation(s)
- Shuling Han
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shiyue Xu
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aijun Qiao
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chan Boriboun
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wenxia Ma
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huadong Li
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dauren Biyashev
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Liu Yang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qinghua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, Hubei, China
| | - Shayi Jiang
- Department of Hematology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 20062, China
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Providence, RI 02908, USA
| | - Prasanna Krishnamurthy
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chunxiang Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stéphane Richard
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Hongyu Qiu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA 30303, USA
| | - Jianyi Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
18
|
Hydrogen sulfide improves vascular repair by promoting endothelial nitric oxide synthase-dependent mobilization of endothelial progenitor cells. J Hypertens 2019; 37:972-984. [DOI: 10.1097/hjh.0000000000001983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Kwon YW, Lee SJ, Heo SC, Lee TW, Park GT, Yoon JW, Kim SC, Shin HJ, Lee SC, Kim JH. Role of CXCR2 in the Ac-PGP-Induced Mobilization of Circulating Angiogenic Cells and its Therapeutic Implications. Stem Cells Transl Med 2018; 8:236-246. [PMID: 30474937 PMCID: PMC6392381 DOI: 10.1002/sctm.18-0035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Circulating angiogenic cells (CACs) have been implicated in the repair of ischemic tissues, and their mobilization from bone marrow is known to be regulated by the activations of chemokine receptors, including CXCR2 and CXCR4. This study was conducted to investigate the role of N‐acetylated proline‐glycine‐proline (Ac‐PGP; a collagen‐derived chemotactic tripeptide) on CAC mobilization and its therapeutic potential for the treatment of peripheral artery diseases. Ac‐PGP was administered daily to a murine hind limb ischemia model, and the effects of Ac‐PGP on blood perfusion and CAC mobilization (Sca1+Flk1+ cells) into peripheral blood were assessed. Intramuscular administration of Ac‐PGP significantly improved ischemic limb perfusion and increased limb salvage rate by increasing blood vessel formation, whereas Ac‐PGP‐induced blood perfusion and angiogenesis in ischemic limbs were not observed in CXCR2‐knockout mice. In addition, Ac‐PGP‐induced CAC mobilization was found to occur in wild‐type mice but not in CXCR2‐knockout mice. Transplantation of bone marrow from green fluorescent protein (GFP) transgenic mice to wild‐type mice showed bone marrow‐derived cells homed to ischemic limbs after Ac‐PGP administration and that GFP‐positive cells contributed to the formation of ILB4‐positive capillaries and α smooth muscle actin (α‐SMA)‐positive arteries. These results suggest CXCR2 activation in bone marrow after Ac‐PGP administration improves blood perfusion and reduces tissue necrosis by inducing CAC mobilization. These findings suggest a new pharmaceutical basis for the treatment of critical limb ischemia. stem cells translational medicine2019;8:236&246
Collapse
Affiliation(s)
- Yang Woo Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seung Jun Lee
- Department of Orthopaedic Surgery, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Soon Chul Heo
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Tae Wook Lee
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Gyu Tae Park
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seung-Chul Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Ho Jin Shin
- Division of Hematology-Oncology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
20
|
Xu S, Tao J, Yang L, Zhang E, Boriboun C, Zhou J, Sun T, Cheng M, Huang K, Shi J, Dong N, Liu Q, Zhao TC, Qiu H, Harris RA, Chandel NS, Losordo DW, Qin G. E2F1 Suppresses Oxidative Metabolism and Endothelial Differentiation of Bone Marrow Progenitor Cells. Circ Res 2018; 122:701-711. [PMID: 29358228 DOI: 10.1161/circresaha.117.311814] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/18/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
RATIONALE The majority of current cardiovascular cell therapy trials use bone marrow progenitor cells (BM PCs) and achieve only modest efficacy; the limited potential of these cells to differentiate into endothelial-lineage cells is one of the major barriers to the success of this promising therapy. We have previously reported that the E2F transcription factor 1 (E2F1) is a repressor of revascularization after ischemic injury. OBJECTIVE We sought to define the role of E2F1 in the regulation of BM PC function. METHODS AND RESULTS Ablation of E2F1 (E2F1 deficient) in mouse BM PCs increases oxidative metabolism and reduces lactate production, resulting in enhanced endothelial differentiation. The metabolic switch in E2F1-deficient BM PCs is mediated by a reduction in the expression of pyruvate dehydrogenase kinase 4 and pyruvate dehydrogenase kinase 2; overexpression of pyruvate dehydrogenase kinase 4 reverses the enhancement of oxidative metabolism and endothelial differentiation. Deletion of E2F1 in the BM increases the amount of PC-derived endothelial cells in the ischemic myocardium, enhances vascular growth, reduces infarct size, and improves cardiac function after myocardial infarction. CONCLUSION Our results suggest a novel mechanism by which E2F1 mediates the metabolic control of BM PC differentiation, and strategies that inhibit E2F1 or enhance oxidative metabolism in BM PCs may improve the effectiveness of cell therapy.
Collapse
Affiliation(s)
- Shiyue Xu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Jun Tao
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Liu Yang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Eric Zhang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Chan Boriboun
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Junlan Zhou
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Tianjiao Sun
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Min Cheng
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Kai Huang
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Jiawei Shi
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Nianguo Dong
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Qinghua Liu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Ting C Zhao
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Hongyu Qiu
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Robert A Harris
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Navdeep S Chandel
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Douglas W Losordo
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.)
| | - Gangjian Qin
- From the Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at Birmingham (S.X., L.Y., E.Z., C.B., G.Q.); Feinberg Cardiovascular Research Institute (S.X., J.Z., T.S., D.W.L., G.Q.) and Department of Medicine - Pulmonary and Critical Care Medicine (N.S.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Hypertension and Vascular Disease, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (S.X., J.T.); Department of Cardiology (L.Y., M.C., K.H.) and Department of Cardiovascular Surgery (J.S., N.D.), Union Hospital of Huazhong University of Science and Technology Tongji Medical College, Wuhan, China; Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China (Q.L.); Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI (T.C.Z.); Department of Basic Science, School of Medicine, Loma Linda University, CA (H.Q.); and Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis (R.A.H.).
| |
Collapse
|
21
|
Prathipati P, Nandi SS, Mishra PK. Stem Cell-Derived Exosomes, Autophagy, Extracellular Matrix Turnover, and miRNAs in Cardiac Regeneration during Stem Cell Therapy. Stem Cell Rev Rep 2017; 13:79-91. [PMID: 27807762 DOI: 10.1007/s12015-016-9696-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell therapy (SCT) raises the hope for cardiac regeneration in ischemic hearts. However, underlying molecular mechanisms for repair of dead myocardium by SCT in the ischemic heart is poorly understood. Growing evidences suggest that cardiac matrix stiffness and differential expressions of miRNAs play a crucial role in stem cell survival and differentiation. However, their roles on transplanted stem cells, for myocardial repair of the ischemic heart, remain unclear. Transplanted stem cells may act in an autocrine and/or paracrine manner to regenerate the dead myocardium. Paracrine mediators such as stem cell-derived exosomes are emerging as a novel therapeutic strategy to overcome some of the limitations of SCT. These exosomes carry microRNAs (miRNAs) that may regulate stem cell differentiation into a specific lineage. MicroRNAs may also contribute to stiffness of surrounding matrix by regulating extracellular matrix (ECM) turnover. The survival of transplanted stem cell depends on its autophagic process that maintains cellular homeostasis. Therefore, exosomes, miRNAs, extracellular matrix turnover, and autophagy may have an integral role in improving the efficacy of SCT. This review elaborates the specific roles of these regulatory components on cardiac regeneration in the ischemic heart during SCT.
Collapse
Affiliation(s)
- Priyanka Prathipati
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shyam Sundar Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Paras Kumar Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
22
|
Wang H, Qiu L, Ma Y, Zhang L, Chen L, Li C, Geng X, You X, Gao X. Naoxintong inhibits myocardial infarction injury by VEGF/eNOS signaling-mediated neovascularization. JOURNAL OF ETHNOPHARMACOLOGY 2017; 209:13-23. [PMID: 28669772 DOI: 10.1016/j.jep.2017.06.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naoxintong capsules (NXT), a traditional Chinese Medical preparation, are widely used for treatment of cardiovascular diseases, while the mechanism is still unclear. MATERIALS AND METHODS Myocardial infarction (MI) was induced by ligation of the left coronary artery in mice. Echocardiographic measurements were performed to do physiological assessments of left ventricle (LV) function. Histological and immunohistochemical staining was used to determine infarct size, capillary density, tissue endothelial nitric oxide synthase (eNOS) expression. Bone Marrow Transplantation (BMT) model and flow cytometric (FCM) analyses were applied to assay endothelial progenitor cells (EPCs) mobilization. Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT-PCR), Western blotting and enzyme-linked immunosorbent assay (ELISA) were performed to detect the expressions of vascular endothelial growth factor (VEGF), kinase domain region (KDR), phosphorylated-Akt (p-Akt), phosphorylated-eNOS (p-eNOS). RESULTS NXT administration reduced myocardium fibrosis and increased myocardium capillary density in response to MI. NXT increased circulating Sca1+/ Fetal liver kinase 1 (Flk1)+ mononuclear cells (MNCs) and soluble Kit ligand (sKitL) of bone marrow (BM) in response to MI. In mice transplanted with green fluorescent protein (GFP) BM cells, NXT increased the numbers of GFP-positive cells at the border zone of the ischemic region in MI-induced mice. NXT increased the numbers of eNOS-expressing BM-derived cells in tissues, which was involved in increased the expressions of VEGF, KDR, p-eNOS, p-Akt in the myocardium. CONCLUSION NXT-mediated recovery in MI-induced mice was involved in mobilization and incorporation of bone marrow-derived EPCs/circulating angiogenic cells (CACs) leading to enhancement of neovascularization via VEGF/eNOS signaling.
Collapse
Affiliation(s)
- Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Lizhen Qiu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yake Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lusha Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunxiao Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Geng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xingyu You
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
23
|
Endothelial Progenitor Cells for Ischemic Stroke: Update on Basic Research and Application. Stem Cells Int 2017; 2017:2193432. [PMID: 28900446 PMCID: PMC5576438 DOI: 10.1155/2017/2193432] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/03/2017] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke is one of the leading causes of human death and disability worldwide. So far, ultra-early thrombolytic therapy is the most effective treatment. However, most patients still live with varying degrees of neurological dysfunction due to its narrow therapeutic time window. It has been confirmed in many studies that endothelial progenitor cells (EPCs), as a kind of adult stem cells, can protect the neurovascular unit by repairing the vascular endothelium and its secretory function, which contribute to the recovery of neurological function after an ischemic stroke. This paper reviews the basic researches and clinical trials of EPCs especially in the field of ischemic stroke and addresses the combination of EPC application with new technologies, including neurovascular intervention, synthetic particles, cytokines, and EPC modification, with the aim of shedding some light on the application of EPCs in treating ischemic stroke in the future.
Collapse
|
24
|
Ronca R, Benkheil M, Mitola S, Struyf S, Liekens S. Tumor angiogenesis revisited: Regulators and clinical implications. Med Res Rev 2017. [PMID: 28643862 DOI: 10.1002/med.21452] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since Judah Folkman hypothesized in 1971 that angiogenesis is required for solid tumor growth, numerous studies have been conducted to unravel the angiogenesis process, analyze its role in primary tumor growth, metastasis and angiogenic diseases, and to develop inhibitors of proangiogenic factors. These studies have led in 2004 to the approval of the first antiangiogenic agent (bevacizumab, a humanized antibody targeting vascular endothelial growth factor) for the treatment of patients with metastatic colorectal cancer. This approval launched great expectations for the use of antiangiogenic therapy for malignant diseases. However, these expectations have not been met and, as knowledge of blood vessel formation accumulates, many of the original paradigms no longer hold. Therefore, the regulators and clinical implications of angiogenesis need to be revisited. In this review, we discuss recently identified angiogenesis mediators and pathways, new concepts that have emerged over the past 10 years, tumor resistance and toxicity associated with the use of currently available antiangiogenic treatment and potentially new targets and/or approaches for malignant and nonmalignant neovascular diseases.
Collapse
Affiliation(s)
- Roberto Ronca
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mohammed Benkheil
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| | - Stefania Mitola
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Sandra Liekens
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| |
Collapse
|
25
|
Zhao H, Shen R, Dong X, Shen Y. Murine Double Minute-2 Inhibition Attenuates Cardiac Dysfunction and Fibrosis by Modulating NF-κB Pathway After Experimental Myocardial Infarction. Inflammation 2017; 40:232-239. [PMID: 27838797 DOI: 10.1007/s10753-016-0473-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inflammation has been implicated in myocardial infarction (MI). MDM2 associates with nuclear factor-κB (NF-κB)-mediated inflammation. However, the role of MDM2 in MI remains unclear. This study aimed to evaluate the impacts of MDM2 inhibition on cardiac dysfunction and fibrosis after experimental MI and the underlying mechanisms. Three-month-old male C57BL/6 mice were subjected to left anterior descending (LAD) coronary artery ligation for induction of myocardial infarction (MI). Immediately after MI induction, mice were treated with Nutlin-3a (100 mg/kg) or vehicle twice daily for 4 weeks. Survival, heart function and fibrosis were assessed. Signaling molecules were detected by Western blotting. Mouse myofibroblasts under oxygen and glucose deprivation were used for in vitro experiments. MDM2 protein expression was significantly elevated in the mouse heart after MI. Compared with vehicle-treated animals, Nutlin-3a treatment reduced the mouse mortality. Nutlin-3a treatment improved heart function and decreased the infarct scar and fibrosis compared with vehicle. Furthermore, MDM2 inhibition restored IκB and inhibited NF-κB activation, leading to suppressed production of proinflammatory cytokines in the heart after MI. The consistent results were obtained in vitro. MDM2 inhibition reduced cardiac dysfunction and fibrosis after MI. These effects of MDM2 inhibition is mediated through modulating NF-κB activation, resulting in inhibition of inflammatory response.
Collapse
Affiliation(s)
- Hao Zhao
- Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ruijuan Shen
- Department of Health, Qingdao Municipal Hospital, No.5 Donghaizhong Road, Qingdao, 266071, Shandong, China.
| | - Xiaobin Dong
- Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yi Shen
- Department of Health, Qingdao Municipal Hospital, No.5 Donghaizhong Road, Qingdao, 266071, Shandong, China
| |
Collapse
|
26
|
Das SK, Yuan YF, Li MQ. An Overview on Current Issues and Challenges of Endothelial Progenitor Cell-Based Neovascularization in Patients with Diabetic Foot Ulcer. Cell Reprogram 2017; 19:75-87. [PMID: 28266867 DOI: 10.1089/cell.2016.0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetic foot ulcer's impaired wound healing, which leads to the development of chronic non-healing wounds and ultimately amputation, is a major problem worldwide. Although recently endothelial progenitor cell-derived cell therapy has been used as a therapeutic intervention to treat diabetic wounds, thereby promoting neovascularization, the results, however, are not satisfactory. In this article, we have discussed the several steps that are involved in the neovascularization process, which might be impaired during diabetes. In addition, we have also discussed the reported possible interventions to correct these impairments. Thus, we have summarized neovascularization as a process with a coordinated sequence of multiple steps and thus, there is the need of a combined therapeutic approach to achieve better treatment outcomes.
Collapse
Affiliation(s)
- Sushant Kumar Das
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University , Shanghai, People's Republic of China
| | - Yi Feng Yuan
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University , Shanghai, People's Republic of China
| | - Mao Quan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University , Shanghai, People's Republic of China
| |
Collapse
|
27
|
Abplanalp WT, Conklin DJ, Cantor JM, Ginsberg MH, Wysoczynski M, Bhatnagar A, O'Toole TE. Enhanced Integrin α4β1-Mediated Adhesion Contributes to a Mobilization Defect of Endothelial Progenitor Cells in Diabetes. Diabetes 2016; 65:3505-3515. [PMID: 27495221 PMCID: PMC5079633 DOI: 10.2337/db16-0634] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
Diabetes is associated with a deficit of circulating endothelial progenitor cells (EPCs), which has been attributed to their defective mobilization from the bone marrow. The basis for this mobilization defect is not completely understood, and we sought to determine if hyperglycemic conditions enhanced EPC adhesion. We found that culturing EPCs in high glucose media increased adhesion to bone marrow stromal cells. This enhanced adhesion was associated with decreased expression of protein kinase A regulatory subunit 1β (PRKAR1β), activation of protein kinase A (PKA), and phosphorylation of α4-integrin on serine 988. This potentiated adhesion was reversed by treatment with a PKA inhibitor, overexpression of PRKAR1β, or expression of a phosphorylation-defective α4-integrin variant (α4[S988A]). Using a model of type 1 diabetes, we showed that α4(S988A)-expressing mice have more circulating EPCs than their wild-type counterparts. Moreover, diabetic α4(S988A) mice demonstrate enhanced revascularization after hind limb ischemia. Thus, we have identified a novel signaling mechanism activating PKA in diabetes (downregulation of an inhibitory regulatory subunit) that leads to deficits of circulating EPCs and impaired vascular repair, which could be reversed by α4-integrin mutation.
Collapse
Affiliation(s)
- Wesley T Abplanalp
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
| | - Joseph M Cantor
- Department of Medicine, University of California, San Diego, San Diego, CA
| | - Mark H Ginsberg
- Department of Medicine, University of California, San Diego, San Diego, CA
| | | | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Timothy E O'Toole
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
| |
Collapse
|
28
|
E2F1 Hinders Skin Wound Healing by Repressing Vascular Endothelial Growth Factor (VEGF) Expression, Neovascularization, and Macrophage Recruitment. PLoS One 2016; 11:e0160411. [PMID: 27490344 PMCID: PMC4973919 DOI: 10.1371/journal.pone.0160411] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/19/2016] [Indexed: 12/21/2022] Open
Abstract
Background Refractory surface of wound and dermal chronic ulcer are largely attributed to poor neovascularization. We have previously shown that E2F1 suppresses VEGF expression in the ischemic heart, and that genetic deletion of E2F1 leads to better cardiac recovery. However, whether E2F1 has a role in dermal wound healing is currently not known. Methods and Results Skin wounds were surgically induced in E2F1-null (E2F1–/–) mice and WT littermates. E2F1–/– displayed an accelerated wound healing including wound closure, dermal thickening and collagen deposition, which was associated with an increased endothelial cell proliferation and greater vessel density in the border zone of the wound. Furthermore, more macrophages were recruited to the skin lesions and the level of VEGF expression was markedly higher in E2F1–/– than in WT mice. Conclusions E2F1 hinders skin wound healing by suppressing VEGF expression, neovascularization, and macrophage recruitment. Strategies that target E2F1 may enhance wound healing.
Collapse
|
29
|
Endothelial progenitor cells promote tumor growth and progression by enhancing new vessel formation. Oncol Lett 2016; 12:793-799. [PMID: 27446353 PMCID: PMC4950911 DOI: 10.3892/ol.2016.4733] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/02/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor growth and progression require new blood vessel formation to deliver nutrients and oxygen for further cell proliferation and to create a neovascular network exit for tumor cell metastasis. Endothelial progenitor cells (EPCs) are a bone marrow (BM)-derived stem cell population that circulates in the peripheral circulation and homes to the tumor bed to participate in new blood vessel formation. In addition to structural support to nascent vessels, these cells can also regulate the angiogenic process by paracrine secretion of a number of proangiogenic growth factors and cytokines, thus playing a crucial role in tumor neovascularization and development. Inhibition of EPC-mediated new vessel formation may be a promising therapeutic strategy in tumor treatment. EPC-mediated neovascularization is a complex process that includes multiple steps and requires a series of cytokines and modulators, thus understanding the underlying mechanisms may provide anti-neovasculogenesis targets that may be blocked for the prevention of tumor development. The present review stresses the process and contribution of EPCs to the formation of new blood vessels in solid tumors, in an attempt to gain an improved understanding of the underlying cellular and molecular mechanisms involved, and to provide a potential effective therapeutic target for cancer treatment.
Collapse
|
30
|
Functional and Biological Role of Endothelial Precursor Cells in Tumour Progression: A New Potential Therapeutic Target in Haematological Malignancies. Stem Cells Int 2015; 2016:7954580. [PMID: 26788072 PMCID: PMC4691637 DOI: 10.1155/2016/7954580] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/19/2015] [Accepted: 08/10/2015] [Indexed: 12/11/2022] Open
Abstract
It was believed that vasculogenesis occurred only during embryo life and that postnatal formation of vessels arose from angiogenesis. Recent findings demonstrate the existence of Endothelial Precursor Cells (EPCs), which take partin postnatal vasculogenesis. EPCs are recruited from the bone marrow under the stimulation of growth factors and cytokines and reach the sites of neovascularization in both physiological and pathological conditions such as malignancies where they contribute to the “angiogenic switch” and tumor progression. An implementation of circulating EPCs in the bloodstream of patients with haematological malignancies has been demonstrated. This increase is strictly related to the bone marrow microvessel density and correlated with a poor prognosis. The EPCs characterization is a very complex process and still under investigation. This literature review aims to provide an overview of the functional and biological role of EPCs in haematological malignancies and to investigate their potential as a new cancer therapeutic target.
Collapse
|
31
|
Particle Radiation-Induced Nontargeted Effects in Bone-Marrow-Derived Endothelial Progenitor Cells. Stem Cells Int 2015; 2015:496512. [PMID: 26074973 PMCID: PMC4436457 DOI: 10.1155/2015/496512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 01/17/2023] Open
Abstract
Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons ((1)H) and high charge and energy (HZE) nuclei (e.g., iron-(56)Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by (1)H- and (56)Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2-15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body (1)H- and (56)Fe-IR mice demonstrated a cyclical (early 5-24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.
Collapse
|
32
|
Cheng M, Huang K, Zhou J, Yan D, Tang YL, Zhao TC, Miller RJ, Kishore R, Losordo DW, Qin G. A critical role of Src family kinase in SDF-1/CXCR4-mediated bone-marrow progenitor cell recruitment to the ischemic heart. J Mol Cell Cardiol 2015; 81:49-53. [PMID: 25655934 DOI: 10.1016/j.yjmcc.2015.01.024] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 01/23/2015] [Accepted: 01/28/2015] [Indexed: 11/17/2022]
Abstract
The G protein-coupled receptor CXCR4 and its ligand stromal-cell derived factor 1 (SDF-1) play a crucial role in directing progenitor cell (PC) homing to ischemic tissue. The Src family protein kinases (SFK) can be activated by, and serve as effectors of, G proteins. In this study we sought to determine whether SFK play a role in SDF-1/CXCR4-mediated PC homing. First, we investigated whether SDF-1/CXCR4 signaling activates SFK. Bone-marrow mononuclear cells (BM MNCs) were isolated from WT and BM-specific CXCR4-KO mice and treated with SDF-1 and/or CXCR4 antagonist AMD3100. SDF-1 treatment rapidly induced phosphorylation (activation) of hematopoietic Src (i.e., Lyn, Fgr, and Hck) in WT cells but not in AMD3100-treated cells or CXCR4-KO cells. Then, we investigated whether SFK are involved in SDF-1/CXCR4-mediated PC chemotaxis. In a combined chemotaxis and endothelial-progenitor-cell (EPC) colony assay, Src inhibitor SU6656 dose-dependently inhibited the SDF-1-induced migration of colony-forming EPCs. Next, we investigated whether SFK play a role in SDF-1/CXCR4-mediated BM PC homing to the ischemic heart. BM MNCs from CXCR4BAC:eGFP reporter mice were i.v. injected into WT and SDF-1BAC:SDF1-RFP transgenic mice following surgically-induced myocardial infarction (MI). eGFP(+) MNCs and eGFP(+)c-kit(+) PCs that were recruited in the infarct border zone in SDF-1BAC:SDF1-RFP recipients were significantly more than that in WT recipients. Treatments of mice with SU6656 significantly reduced eGFP(+) and eGFP(+)c-kit(+) cell recruitment in both WT and SDF-1BAC:RFP recipients and abrogated the difference between the two groups. Remarkably, PCs isolated from BM-specific C-terminal Src kinase (CSK)-KO (Src activated) mice were recruited more efficiently than PCs from WT PCs in the WT recipients. In conclusion, SFK are activated by SDF-1/CXCR4 signaling and play an essential role in SDF-1/CXCR4-mediated BM PC chemotactic response and ischemic cardiac recruitment.
Collapse
Affiliation(s)
- Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dewen Yan
- Department of Endocrinology, The 2nd Renmin Hospital, Shenzhen, PR China
| | - Yao-Liang Tang
- Vascular Biology Center, Department of Medicine, Medical College of Georgia/Georgia Regents University, Augusta, GA, USA
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Douglas W Losordo
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
33
|
Tang JM, Luo B, Xiao JH, Lv YX, Li XL, Zhao JH, Zheng F, Zhang L, Chen L, Yang JY, Guo LY, Wang L, Yan YW, Pan YM, Wang JN, Li DS, Wan Y, Chen SY. VEGF-A promotes cardiac stem cell engraftment and myocardial repair in the infarcted heart. Int J Cardiol 2015; 183:221-31. [PMID: 25679991 DOI: 10.1016/j.ijcard.2015.01.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/24/2014] [Accepted: 01/25/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND The objective of this study was to determine whether vascular endothelial growth factor (VEGF)-A subtypes improve cardiac stem cell (CSC) engraftment and promote CSC-mediated myocardial repair in the infarcted heart. METHODS CSCs were treated with VEGF receptor (VEGFR) inhibitors, VCAM-1 antibody (VCAM-1-Ab), or PKC-α inhibitor followed by the treatment with VEGF-A. CSC adhesion assays were performed in vitro. In vivo, the PKH26-labeled and VCAM-1-Ab or PKC-α inhibitor pre-treated CSCs were treated with VEGF-A followed by implantation into infarcted rat hearts. The hearts were then collected for measuring CSC engraftment and evaluating cardiac fibrosis and function 3 or 28days after the CSC transplantation. RESULTS All three VEGF-A subtypes promoted CSC adhesion to extracellular matrix and endothelial cells. VEGF-A-mediated CSC adhesion required VEGFR and PKCα signaling. Importantly, VEGF-A induced VCAM-1, but not ICAM-1 expression in CSCs through PKCα signaling. In vivo, VEGF-A promoted the engraftment of CSCs in infarcted hearts, which was attenuated by PKCα inhibitor or VCAM-1-Ab. Moreover, VEGF-A-mediated CSC engraftment resulted in a reduction in infarct size and fibrosis. Functional studies showed that the transplantation of the VEGF-A-treated CSCs stimulated extensive angiomyogenesis in infarcted hearts as indicated by the expression of cardiac troponin T and von Willebrand factor, leading to an improved performance of left ventricle. Blockade of PKCα signaling or VCAM-1 significantly diminished the beneficial effects of CSCs treated with VEGF-A. CONCLUSION VEGF-A promotes myocardial repair through, at least in part, enhancing the engraftment of CSCs mediated by PKCα/VCAM-1 pathway.
Collapse
Affiliation(s)
- Jun-Ming Tang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China; Center for Medical Research and Department of Physiology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, China.
| | - Bin Luo
- Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China
| | - Jun-hui Xiao
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yan-xia Lv
- Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China
| | - Xiao-lin Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jin-he Zhao
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lei Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Long Chen
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jian-Ye Yang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lin-Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yu-Wen Yan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Ya-Mo Pan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jia-Ning Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Dong-sheng Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yu Wan
- Center for Medical Research and Department of Physiology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, China.
| | - Shi-You Chen
- Department of Physiology & Pharmacology, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
34
|
Lu A, Wang L, Qian L. The role of eNOS in the migration and proliferation of bone-marrow derived endothelial progenitor cells and in vitro angiogenesis. Cell Biol Int 2015; 39:484-90. [PMID: 25492215 DOI: 10.1002/cbin.10405] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/28/2014] [Indexed: 12/23/2022]
Abstract
The role of endothelial nitric oxide synthase (eNOS) in the activities of endothelial progenitor cells (EPCs) including migration, proliferation, and tube formation in vitro was investigated. EPCs were obtained from rat bone mononuclear cells by culturing for 7-10 days in EGM-2MV and identified by their capacity for FITC-UEA-1 binding and acetylated low-density lipoprotein (Dil-ac-LDL) intake using fluorescence microscopy. Migration, proliferation and tube formation activities were assessed in the presence or absence of N(ω)-nitro-L-argininemethylester (L-NAME), an eNOS inhibitor. mRNA and protein expression of CXCR4, CXCR7, VEGFR2, and eNOS were detected by real-time PCR and western blotting in the presence or absence of L-NAME. Nitric oxide production was detected by nitrate reductase in the presence or absence of L-NAME. Typical spindle-shaped cells appeared on the 7(th)-10(th) day and confluence reached about 80%. The percentage of FITC-UEA-1 and Dil-ac-LDL double-stained cells was about 85%. Cell migration, proliferation, and tube formation were significantly weakened after eNOS was inhibited (P < 0.05), and the expressions of CXCR4 and eNOS were significantly reduced (P < 0.05, respectively), but there was little change in CXCR7 and VEGFR2. NO production was dramatically decreased after eNOS was inhibited (P < 0.05). In summary, L-NAME significantly reduced the expression of eNOS and NO production by EPCs and inhibited migration, proliferation and tube formation by these cells, suggesting that eNOS affects EPC activities; CXCR4 may be implicated in the action of eNOS.
Collapse
Affiliation(s)
- Aizhen Lu
- Departments of Pediatrics, Children's Hospital of Fudan University, Shanghai, 201102, P. R. China
| | | | | |
Collapse
|
35
|
Wang P, Du H, Zhou CC, Song J, Liu X, Cao X, Mehta JL, Shi Y, Su DF, Miao CY. Intracellular NAMPT-NAD+-SIRT1 cascade improves post-ischaemic vascular repair by modulating Notch signalling in endothelial progenitors. Cardiovasc Res 2014; 104:477-488. [PMID: 25341895 DOI: 10.1093/cvr/cvu220] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS Intracellular nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme for nicotinamide adenine dinucleotide (NAD(+)) biosynthesis. This study investigated the role of NAMPT-mediated NAD(+) signalling in post-ischaemic vascular repair. METHODS AND RESULTS Mouse hind-limb ischaemia up-regulated NAMPT expression and NAD(+) level in bone marrow (BM). Pharmacological inhibition of NAMPT by a chemical inhibitor FK866 impaired the mobilization of endothelial progenitor cells (EPCs) from BM upon ischaemic stress. Transgenic mice overexpressing NAMPT (Tg mice), but not H247A-mutant dominant-negative NAMPT (DN-Tg mice), exhibited enhanced capillary density, increased number of proliferating endothelial cells, improved blood flow recovery, and augmented collateral arterioles in the ischaemic limb. In cultured BM-derived EPCs, inhibition of NAMPT suppressed proliferation, migration, and tube formation, whereas overexpression of NAMPT induced opposite effects. The promoting effects of NAMPT on EPCs were abolished by silencing of sirtuin 1 (SIRT1), rather than silencing of SIRT2-7. Overexpression of NAMPT led to a SIRT1-depedent enhancement of Notch-1 intracellular domain deacetylation, which inhibited Delta-like ligand-4 (DLL4)-Notch signalling and thereby up-regulated of VEGFR-2 and VEGFR-3. Injection of recombinant VEGF induced a more pronounced EPC mobilization in Tg, but not in DN-Tg, mice. Furthermore, overexpression of NAMPT down-regulated Fringe family glycosyltransferases in a SIRT1-dependent manner, which rendered Notch more sensitive to the pro-angiogenic ligand Jagged1 rather than the anti-angiogenic ligand DLL4. CONCLUSIONS These results demonstrate that intracellular NAMPT-NAD(+)-SIRT1 cascade improves post-ischaemic neovascularization. The modulation of Notch signalling may contribute to the enhanced post-ischaemic neovascularization.
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China
| | - Hui Du
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China Department of Pharmacy, General Hospital of Lanzhou Military Region, Lanzhou, China
| | - Can-Can Zhou
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China
| | - Jie Song
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China
| | - Xingguang Liu
- Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xuetao Cao
- Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Jawahar L Mehta
- Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yi Shi
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, 325 Guo He Road, Shanghai 200433, China
| |
Collapse
|
36
|
Wu M, Zhou J, Cheng M, Boriboun C, Biyashev D, Wang H, Mackie A, Thorne T, Chou J, Wu Y, Chen Z, Liu Q, Yan H, Yang Y, Jie C, Tang YL, Zhao TC, Taylor RN, Kishore R, Losordo DW, Qin G. E2F1 suppresses cardiac neovascularization by down-regulating VEGF and PlGF expression. Cardiovasc Res 2014; 104:412-22. [PMID: 25341896 DOI: 10.1093/cvr/cvu222] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS The E2F transcription factors are best characterized for their roles in cell-cycle regulation, cell growth, and cell death. Here we investigated the potential role of E2F1 in cardiac neovascularization. METHODS AND RESULTS We induced myocardial infarction (MI) by ligating the left anterior descending artery in wild-type (WT) and E2F1(-/-) mice. E2F1(-/-) mice demonstrated a significantly better cardiac function and smaller infarct sizes than WT mice. At infarct border zone, capillary density and endothelial cell (EC) proliferation were greater, apoptotic ECs were fewer, levels of VEGF and placental growth factor (PlGF) were higher, and p53 level was lower in E2F1(-/-) than in WT mice. Blockade of VEGF receptor 2 (VEGFR2) signalling with the selective inhibitor SU5416 or with the VEGFR2-blocking antibody DC101 abolished the differences between E2F1(-/-) mice and WT mice in cardiac function, infarct size, capillary density, EC proliferation, and EC apoptosis. In vitro, hypoxia-induced VEGF and PlGF up-regulation was significantly greater in E2F1(-/-) than in WT cardiac fibroblasts, and E2F1 overexpression suppressed PlGF up-regulation in both WT and p53(-/-) cells; however, VEGF up-regulation was suppressed only in WT cells. E2F1 interacted with and stabilized p53 under hypoxic conditions, and both E2F1 : p53 binding and the E2F1-induced suppression of VEGF promoter activity were absent in cells that expressed an N-terminally truncated E2F1 mutant. CONCLUSION E2F1 limits cardiac neovascularization and functional recovery after MI by suppressing VEGF and PlGF up-regulation through p53-dependent and -independent mechanisms, respectively.
Collapse
Affiliation(s)
- Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Junlan Zhou
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chan Boriboun
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Dauren Biyashev
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Hong Wang
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Alexander Mackie
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Tina Thorne
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Jonathan Chou
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhishui Chen
- Organ Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qinghua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China
| | - Hongbin Yan
- Cardiology Department, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya Yang
- Department of Echocardiography, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Chunfa Jie
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yao-Liang Tang
- Department of Medicine, Vascular Biology Center, Medical College of Georgia/Georgia Regents University, Augusta, GA, USA
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI, USA
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Douglas W Losordo
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Gangjian Qin
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| |
Collapse
|
37
|
Hou X, Zeng H, He X, Chen JX. Sirt3 is essential for apelin-induced angiogenesis in post-myocardial infarction of diabetes. J Cell Mol Med 2014; 19:53-61. [PMID: 25311234 PMCID: PMC4288349 DOI: 10.1111/jcmm.12453] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/05/2014] [Indexed: 12/28/2022] Open
Abstract
Heart failure following myocardial infarction (MI) is the leading cause of death in diabetic patients. Angiogenesis contributes to cardiac repair and functional recovery in post-MI. Our previous study shows that apelin (APLN) increases Sirtuin 3 (Sirt3) expression and ameliorates diabetic cardiomyopathy. In this study, we further investigated the direct role of Sirt3 in APLN-induced angiogenesis in post-MI model of diabetes. Wild-type (WT) and Sirt3 knockout (Sirt3KO) mice were induced into diabetes by i.p. streptozotocin (STZ). STZ mice were then subjected to MI followed by immediate intramyocardial injection with adenovirus-apelin (Ad-APLN). Our studies showed that Sirt3 expression was significantly reduced in the hearts of STZ mice. Ad-APLN treatment resulted in up-regulation of Sirt3, angiopoietins/Tie-2 and VEGF/VEGFR2 expression together with increased myocardial vascular densities in WT-STZ+MI mice, but these alterations were not observed in Sirt3KO-STZ+MI mice. In vitro, overexpression of APLN increased Sirt3 expression and angiogenesis in endothelial progenitor cells (EPC) from WT mice, but not in EPC from Sirt3KO mice. APLN gene therapy increases angiogenesis and improves cardiac functional recovery in diabetic hearts via up-regulation of Sirt3 pathway.
Collapse
Affiliation(s)
- Xuwei Hou
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | |
Collapse
|
38
|
A novel molecule Me6TREN promotes angiogenesis via enhancing endothelial progenitor cell mobilization and recruitment. Sci Rep 2014; 4:6222. [PMID: 25164363 PMCID: PMC5385830 DOI: 10.1038/srep06222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/11/2014] [Indexed: 12/29/2022] Open
Abstract
Critical limb ischaemia is the most severe clinical manifestation of peripheral arterial disease. The circulating endothelial progenitor cells (EPCs) play important roles in angiogenesis and ischemic tissue repair. The increase of circulating EPC numbers by using mobilization agents is critical for obtaining a better therapeutic outcome in patients with ischemic disease. Here, we firstly report a novel small molecule, Me6TREN (Me6), can efficiently mobilize EPCs into the blood circulation. Single injection of Me6 induced a long-lasting increase in circulating Flk-1+ Sca-1+ EPC numbers. In a mouse hind limb ischemia (HLI) model, local intramuscular transplantation of these Me6-mobilized cells accelerated the blood flow restoration in the ischemic muscles. More importantly, systemic administration of Me6 notably increased the capillary density, arteriole density and regenerative muscle weight in the ischemic tissue of HLI. Mechanistically, we found Me6 reduced stromal cell-derived factor-1α level in bone marrow by up-regulation of matrix metallopeptidase-9 expression, which allowed the dissemination of EPCs into peripheral blood. These data indicate that Me6 may represent a potentially useful therapy for ischemic disease via enhancing autologous EPC recruitment and promote angiogenesis.
Collapse
|
39
|
Abstract
Myocardial infarction is a leading cause of death among all cardiovascular diseases. The analysis of molecular mechanisms by which the ischemic myocardium initiates repair and remodeling indicates that secreted soluble factors are key players in communication to local and distant tissues, such as bone marrow. Recently, actively secreted membrane vesicles, including exosomes, are being recognized as new candidates with important roles in intercellular and tissue-level communication. In this review, we critically examine the emerging role of exosomes in local and distant microcommunication mechanisms after myocardial infarction. A comprehensive understanding of the role of exosomes in cardiac repair after myocardial infarction could bridge a major gap in knowledge of the repair mechanism after myocardial injury.
Collapse
Affiliation(s)
- Susmita Sahoo
- From Feinberg Cardiovascular Research Institute, Chicago, IL (S.S., D.W.L.); Northwestern Memorial Hospital, Chicago, IL (D.W.L.); and NeoStem, Inc, New York, NY (D.W.L.)
| | | |
Collapse
|
40
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
41
|
Makridakis M, Roubelakis MG, Vlahou A. Stem cells: Insights into the secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2380-4. [DOI: 10.1016/j.bbapap.2013.01.032] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/19/2013] [Accepted: 01/23/2013] [Indexed: 01/06/2023]
|
42
|
Yao L, Heuser-Baker J, Herlea-Pana O, Iida R, Wang Q, Zou MH, Barlic-Dicen J. Bone marrow endothelial progenitors augment atherosclerotic plaque regression in a mouse model of plasma lipid lowering. Stem Cells 2013; 30:2720-31. [PMID: 23081735 DOI: 10.1002/stem.1256] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 09/14/2012] [Indexed: 12/30/2022]
Abstract
The major event initiating atherosclerosis is hypercholesterolemia-induced disruption of vascular endothelium integrity. In settings of endothelial damage, endothelial progenitor cells (EPCs) are mobilized from bone marrow into circulation and home to sites of vascular injury where they aid endothelial regeneration. Given the beneficial effects of EPCs in vascular repair, we hypothesized that these cells play a pivotal role in atherosclerosis regression. We tested our hypothesis in the atherosclerosis-prone mouse model in which hypercholesterolemia, one of the main factors affecting EPC homeostasis, is reversible (Reversa mice). In these mice, normalization of plasma lipids decreased atherosclerotic burden; however, plaque regression was incomplete. To explore whether endothelial progenitors contribute to atherosclerosis regression, bone marrow EPCs from a transgenic strain expressing green fluorescent protein (GFP) under the control of endothelial cell-specific Tie2 promoter (Tie2-GFP(+)) were isolated. These cells were then adoptively transferred into atheroregressing Reversa recipients where they augmented plaque regression induced by reversal of hypercholesterolemia. Advanced plaque regression correlated with engraftment of Tie2-GFP(+) EPCs into endothelium and resulted in an increase in atheroprotective nitric oxide and improved vascular relaxation. Similarly augmented plaque regression was also detected in regressing Reversa mice treated with the stem cell mobilizer AMD3100 which also mobilizes EPCs to peripheral blood. We conclude that correction of hypercholesterolemia in Reversa mice leads to partial plaque regression that can be augmented by AMD3100 treatment or by adoptive transfer of EPCs. This suggests that direct cell therapy or indirect progenitor cell mobilization therapy may be used in combination with statins to treat atherosclerosis.
Collapse
Affiliation(s)
- Longbiao Yao
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Balaji S, King A, Crombleholme TM, Keswani SG. The Role of Endothelial Progenitor Cells in Postnatal Vasculogenesis: Implications for Therapeutic Neovascularization and Wound Healing. Adv Wound Care (New Rochelle) 2013; 2:283-295. [PMID: 24527350 DOI: 10.1089/wound.2012.0398] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Indexed: 01/16/2023] Open
Abstract
SIGNIFICANCE Postnatal vasculogenesis mediated via endothelial progenitor cells (EPCs) contributes to re-endothelialization and augments neovascularization after ischemia and tissue injury, providing a novel therapeutic application. However, controversy exists with respect to the origin, identification, and contributions of the EPCs to neovascularization, necessitating further study. RECENT ADVANCES Bone marrow (BM) or circulating cells expressing cd133/vascular endothelial growth factor receptor 2 include those with endothelial progenitor capacity. Increasing evidence suggests that there are additional BM-derived (myeloid; mesenchymal cells) and non-BM-derived (peripheral and cord-blood; tissue-resident) cell populations which also give rise to endothelial cells (ECs) and contribute to re-endothelialization and growth factor release after ischemia and tissue injury. Currently, EPCs are being used as diagnostic markers for the assessment of cardiovascular and tumor risk/progression. Techniques aimed at enhancing ex vivo expansion and the therapeutic potential of these cells are being optimized. CRITICAL ISSUES Mobilization and EPC-mediated neovascularization are critically regulated. Stimulatory (growth factors, statins, and exercise) or inhibitory factors (obesity, diabetes, and other cardiovascular diseases) modulate EPC numbers and function. Recruitment and incorporation of EPCs require a coordinated sequence of signaling events, including adhesion, migration (by integrins), and chemoattraction. Finally, EPCs differentiate into ECs and/or secrete angiogenic growth factors. These cells are highly plastic, and depending on the microenvironment and presence of other cells, EPCs transdifferentiate and/or undergo cell fusion and become cells of a different lineage. Therefore, in vitro culture conditions should be optimized to mimic the in vivo milieu to fully characterize the biological function and contribution of EPCs to postnatal vasculogenesis. FUTURE DIRECTIONS Advances in characterization of the EPC biology and enhancement of EPC functions are required. In addition, innovative tissue-engineered carrier matrices that permit embedding of EPCs and provide optimal conditions for EPC survival and endothelial outgrowth will further contribute to EPC-mediated therapeutic applications in wound healing and ischemia repair.
Collapse
Affiliation(s)
- Swathi Balaji
- Center for Molecular Fetal Therapy, Division of Pediatric, General, Thoracic, and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Alice King
- Center for Molecular Fetal Therapy, Division of Pediatric, General, Thoracic, and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Timothy M. Crombleholme
- Center for Molecular Fetal Therapy, Division of Pediatric, General, Thoracic, and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Children's Surgery, Children's Hospital Colorado and the University of Colorado School of Medicine, Aurora, Colorado
| | - Sundeep G. Keswani
- Center for Molecular Fetal Therapy, Division of Pediatric, General, Thoracic, and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
44
|
Contrasting roles of E2F2 and E2F3 in cardiac neovascularization. PLoS One 2013; 8:e65755. [PMID: 23799044 PMCID: PMC3683051 DOI: 10.1371/journal.pone.0065755] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/29/2013] [Indexed: 01/23/2023] Open
Abstract
Insufficient neovascularization, characterized by poor endothelial cell (EC) growth, contributes to the pathogenesis of ischemic heart disease and limits cardiac tissue preservation and regeneration. The E2F family of transcription factors are critical regulators of the genes responsible for cell-cycle progression and growth; however, the specific roles of individual E2Fs in ECs are not well understood. Here we investigated the roles of E2F2 and E2F3 in EC growth, angiogenesis, and their functional impact on myocardial infarction (MI). An endothelial-specific E2F3-deficient mouse strain VE-Cre; E2F3(fl/fl) was generated, and MI was surgically induced in VE-Cre; E2F3(fl/fl) and E2F2-null (E2F2 KO) mice and their wild-type (WT) littermates, VE-Cre; E2F3(+/+) and E2F2 WT, respectively. The cardiac function, infarct size, and vascular density were significantly better in E2F2 KO mice and significantly worse in VE-Cre; E2F3(fl/fl) mice than in their WT littermates. The loss of E2F2 expression was associated with an increase in the proliferation of ECs both in vivo and in vitro, while the loss of E2F3 expression led to declines in EC proliferation. Thus, E2F3 promotes while E2F2 suppresses ischemic cardiac repair through corresponding changes in EC proliferation; and differential targeting of specific E2F members may provide a novel strategy for therapeutic angiogenesis of ischemic heart disease.
Collapse
|
45
|
Zhou J, Cheng M, Liao YH, Hu Y, Wu M, Wang Q, Qin B, Wang H, Zhu Y, Gao XM, Goukassian D, Zhao TC, Tang YL, Kishore R, Qin G. Rosuvastatin enhances angiogenesis via eNOS-dependent mobilization of endothelial progenitor cells. PLoS One 2013; 8:e63126. [PMID: 23704894 PMCID: PMC3660394 DOI: 10.1371/journal.pone.0063126] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/29/2013] [Indexed: 01/08/2023] Open
Abstract
Circulating endothelial progenitor cells (circEPCs) of bone marrow (BM) origin contribute to postnatal neovascularization and represent a potential therapeutic target for ischemic disease. Statins are beneficial for ischemia disease and have been implicated to increase neovascularization via mechanisms independent of lipid lowering. However, the effect of Statins on EPC function is not completely understood. Here we sought to investigate the effects of Rosuvastatin (Ros) on EPC mobilization and EPC-mediated neovascularization during ischemic injury. In a mouse model of surgically-induced hindlimb ischemia (HLI), treatment of mice with low dose (0.1 mg/kg) but not high dose (5 mg/kg) significantly increased capillary density and accelerated blood flow recovery, as compared to saline-treated group. When HLI was induced in mice that had received Tie2/LacZ BM transplantation, Ros treatment led a significantly larger amount of endothelial cells (ECs) of BM origin incorporated at ischemic sites than saline. After treatment of mice with a single low dose of Ros, circEPCs significantly increased from 2 h, peaked at 4 h, declined until 8 h. In a growth-factor reduced Matrigel plug-in assay, Ros treatment for 5 d induced endothelial lineage differentiation in vivo. Interestingly, the enhanced circEPCs and post-HLI neovascularization stimulated by Ros were blunted in mice deficient in endothelial nitric oxide synthase (eNOS), and Ros increased p-Akt/p-eNOS levels in EPCs in vitro, indicating these effects of Ros are dependent on eNOS activity. We conclude that Ros increases circEPCs and promotes their de novo differentiation through eNOS pathway.
Collapse
Affiliation(s)
- Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Min Cheng
- Department of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yu-Hua Liao
- Department of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Bo Qin
- Weinberg College of Arts and Sciences, Northwestern, Chicago, Illinois, United States of America
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Xiu-Mei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - David Goukassian
- CardioVascular Systems Biology, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ting C. Zhao
- Department of Surgery, Boston University Medical School, Roger William Medical Center, Providence, Rhode Island, United States of America
| | - Yao-Liang Tang
- Division of Cardiovascular Disease, Cardiovascular Research Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Raj Kishore
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
46
|
de la Puente P, Muz B, Azab F, Azab AK. Cell trafficking of endothelial progenitor cells in tumor progression. Clin Cancer Res 2013; 19:3360-8. [PMID: 23665736 DOI: 10.1158/1078-0432.ccr-13-0462] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Blood vessel formation plays an essential role in many physiologic and pathologic processes, including normal tissue growth and healing, as well as tumor progression. Endothelial progenitor cells (EPC) are a subtype of stem cells with high proliferative potential that are capable of differentiating into mature endothelial cells, thus contributing to neovascularization in tumors. In response to tumor-secreted cytokines, EPCs mobilize from the bone marrow to the peripheral blood, home to the tumor site, and differentiate to mature endothelial cells and secrete proangiogenic factors to facilitate vascularization of tumors. In this review, we summarize the expression of surface markers, cytokines, receptors, adhesion molecules, proteases, and cell signaling mechanisms involved in the different steps (mobilization, homing, and differentiation) of EPC trafficking from the bone marrow to the tumor site. Understanding the biologic mechanisms of EPC cell trafficking opens a window for new therapeutic targets in cancer.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri 63108, USA
| | | | | | | |
Collapse
|
47
|
Zhou J, Cheng M, Wu M, Boriboun C, Jujo K, Xu S, Zhao TC, Tang YL, Kishore R, Qin G. Contrasting roles of E2F2 and E2F3 in endothelial cell growth and ischemic angiogenesis. J Mol Cell Cardiol 2013; 60:68-71. [PMID: 23603666 DOI: 10.1016/j.yjmcc.2013.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/28/2013] [Accepted: 04/10/2013] [Indexed: 01/03/2023]
Abstract
The growth of new blood vessels after ischemic injury requires endothelial cells (ECs) to divide and proliferate, and the E2F transcription factors are key regulators of the genes responsible for cell-cycle progression; however, the specific roles of individual E2Fs in ECs are largely unknown. To determine the roles of E2F2 and E2F3 in EC proliferation and the angiogenic response to ischemic injury, hind-limb ischemia was surgically induced in E2F2(-/-) mice, endothelial-specific E2F3-knockout (EndoE2F3(∆/∆)) mice, and their littermates with wild-type E2F2 and E2F3 expression. Two weeks later, Laser-Doppler perfusion measurements, capillary density, and endothelial proliferation were significantly greater in E2F2(-/-) mice and significantly lower in EndoE2F3(∆/∆) mice than in their littermates, and EndoE2F3(∆/∆) mice also developed toe and limb necrosis. The loss of E2F2 expression was associated with increases in the proliferation and G1/S-phase gene expression of isolated ECs, while the loss of E2F3 expression led to declines in these parameters. Thus E2F2 impairs, and endothelial E2F3 promotes, the angiogenic response to peripheral ischemic injury through corresponding changes in EC cell-cycle progression.
Collapse
Affiliation(s)
- Junlan Zhou
- Feinberg Cardiovascular Research Institute, Department of Medicine-Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Li DW, Liu ZQ, Wei J, Liu Y, Hu LS. Contribution of endothelial progenitor cells to neovascularization (Review). Int J Mol Med 2012; 30:1000-6. [PMID: 22922670 DOI: 10.3892/ijmm.2012.1108] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/30/2012] [Indexed: 11/05/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a cell population mobilized from bone marrow into the peripheral circulation and recruited into sites of vessel injury to participate in blood vessel formation in both physiological and pathological conditions. Due to the lack of unique surface markers and different isolation methods, EPCs represent heterogeneous cell populations including cells of myeloid or endothelial origin. Evidence suggests that EPCs play a critical role in postnatal blood vessel formation and vascular homeostasis and provide a promising therapy for vascular disease. However, the mechanisms by which EPCs participate in new vessel formation are still incompletely understood. We review the process of EPCs in neovascularization including EPC mobilization, migration, adhesion and effect on new vessel formation, in an attempt to better understand the underlying mechanisms and to provide potential effective management for the treatment of patients with vascular disease.
Collapse
Affiliation(s)
- Da-Wei Li
- Department of Neurology, The First Hospital of Jilin University, Changchun, PR China
| | | | | | | | | |
Collapse
|
49
|
Caiado F, Dias S. Endothelial progenitor cells and integrins: adhesive needs. FIBROGENESIS & TISSUE REPAIR 2012; 5:4. [PMID: 22410175 PMCID: PMC3323425 DOI: 10.1186/1755-1536-5-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/12/2012] [Indexed: 02/07/2023]
Abstract
In the last decade there have been multiple studies concerning the contribution of endothelial progenitor cells (EPCs) to new vessel formation in different physiological and pathological settings. The process by which EPCs contribute to new vessel formation in adults is termed postnatal vasculogenesis and occurs via four inter-related steps. They must respond to chemoattractant signals and mobilize from the bone marrow to the peripheral blood; home in on sites of new vessel formation; invade and migrate at the same sites; and differentiate into mature endothelial cells (ECs) and/or regulate pre-existing ECs via paracrine or juxtacrine signals. During these four steps, EPCs interact with different physiological compartments, namely bone marrow, peripheral blood, blood vessels and homing tissues. The success of each step depends on the ability of EPCs to interact, adapt and respond to multiple molecular cues. The present review summarizes the interactions between integrins expressed by EPCs and their ligands: extracellular matrix components and cell surface proteins present at sites of postnatal vasculogenesis. The data summarized here indicate that integrins represent a major molecular determinant of EPC function, with different integrin subunits regulating different steps of EPC biology. Specifically, integrin α4β1 is a key regulator of EPC retention and/or mobilization from the bone marrow, while integrins α5β1, α6β1, αvβ3 and αvβ5 are major determinants of EPC homing, invasion, differentiation and paracrine factor production. β2 integrins are the major regulators of EPC transendothelial migration. The relevance of integrins in EPC biology is also demonstrated by many studies that use extracellular matrix-based scaffolds as a clinical tool to improve the vasculogenic functions of EPCs. We propose that targeted and tissue-specific manipulation of EPC integrin-mediated interactions may be crucial to further improve the usage of this cell population as a relevant clinical agent.
Collapse
Affiliation(s)
- Francisco Caiado
- Angiogenesis Laboratory, CIPM, Instituto Português de Oncologia Francisco Gentil, EPE, Lisboa, Portugal.
| | | |
Collapse
|
50
|
Park JH, Yoon JY, Ko SM, Jin SA, Kim JH, Cho CH, Kim JM, Lee JH, Choi SW, Seong IW, Jeong JO. Endothelial progenitor cell transplantation decreases lymphangiogenesis and adverse myocardial remodeling in a mouse model of acute myocardial infarction. Exp Mol Med 2012; 43:479-85. [PMID: 21694495 DOI: 10.3858/emm.2011.43.8.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cardiac lymphatic system in the remodeling after acute myocardial infarction (AMI) has been overlooked. We wanted to investigate the role of bone marrow-derived endothelial progenitor cells (EPCs) and their contribution to lymphatic distribution in myocardial remodeling after AMI. Mouse (C57bl/6J) MI models were created by ligation of the left anterior descending coronary artery and were treated with phosphate buffered saline (PBS) or EPCs. Real-time RT-PCR with 2- to 4-week myocardial tissue samples revealed that lymphangiogenetic factors such as vascular endothelial growth factor (VEGF)-C (8.5 fold, P < 0.05), VEGF-D (6.1 fold, P < 0.05), Lyve-1 (15 fold, P < 0.05), and Prox-1 (11 fold, P < 0.05) were expressed at significantly higher levels in the PBS group than the EPC group. The PBS group also showed a significantly higher density of lymphatic vessels in the peri-infarction area. Echocardiography showed that from 2 weeks after the treatment, left ventricle (LV) dimensions at both systole and diastole were significantly smaller in the EPC group than in the PBS group (P < 0.01) and LV fractional shortening was higher in the EPC group accordingly (P < 0.01). Lymphangiogenic markers increased in a mouse MI model. EPC transplantation decreased lymphangiogenesis and adverse ventricular remodeling after AMI. These novel findings suggest that new lymphatic vessels may be formed in severely damaged myocardium, and may be involved in adverse myocardial remodeling after AMI.
Collapse
Affiliation(s)
- Jae-Hyeong Park
- Division of Cardiology, Chungnam National University, Daejeon 301-721, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|