1
|
Lee YS, Choi JR, Kim JB. Gene Therapy for Cardiovascular Disease: Clinical Perspectives. Yonsei Med J 2024; 65:557-571. [PMID: 39313446 PMCID: PMC11427124 DOI: 10.3349/ymj.2024.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of death in the United States, with its prevalence steadily on the rise. Traditional therapeutic approaches, such as pharmacological treatment, cardiovascular intervention, and surgery, have inherent limitations. In response to these challenges, cardiac gene therapy has emerged as a promising alternative for treating CVD patients. However, several obstacles persist, including the low efficiency of gene transduction, immune reactions to vectors or transduced cells, and the occurrence of off-target effects. While preclinical research has demonstrated significant success in various CVD model in both small and large animals, the translation of these findings to clinical applications has, for the most part, yielded disappointing results, except for some early, albeit small, trials. This review aims to provide a comprehensive summary of recent preclinical and clinical studies on gene therapy for various CVDs. Additionally, we discuss the existing limitations and challenges that hinder the widespread clinical application of cardiac gene therapy.
Collapse
Affiliation(s)
- Young Shin Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Ran Choi
- College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
- College of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
2
|
Liu T, Zhang J, Chang F, Sun M, He J, Ai D. Role of endothelial Raptor in abnormal arteriogenesis after lower limb ischaemia in type 2 diabetes. Cardiovasc Res 2024; 120:1218-1234. [PMID: 38722901 DOI: 10.1093/cvr/cvae105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 09/03/2024] Open
Abstract
AIMS Proper arteriogenesis after tissue ischaemia is necessary to rebuild stable blood circulation; nevertheless, this process is impaired in type 2 diabetes mellitus (T2DM). Raptor is a scaffold protein and a component of mammalian target of rapamycin complex 1 (mTORC1). However, the role of the endothelial Raptor in arteriogenesis under the conditions of T2DM remains unknown. This study investigated the role of endothelial Raptor in ischaemia-induced arteriogenesis during T2DM. METHODS AND RESULTS Although endothelial mTORC1 is hyperactive in T2DM, we observed a marked reduction in the expression of endothelial Raptor in two mouse models and in human vessels. Inducible endothelial-specific Raptor knockout severely exacerbated impaired hindlimb perfusion and arteriogenesis after hindlimb ischaemic injury in 12-week high-fat diet fed mice. Additionally, we found that Raptor deficiency dampened vascular endothelial growth factor receptor 2 (VEGFR2) signalling in endothelial cells (ECs) and inhibited VEGF-induced cell migration and tube formation in a PTP1B-dependent manner. Furthermore, mass spectrometry analysis indicated that Raptor interacts with neuropilin 1 (NRP1), the co-receptor of VEGFR2, and mediates VEGFR2 trafficking by facilitating the interaction between NRP1 and Synectin. Finally, we found that EC-specific overexpression of the Raptor mutant (loss of mTOR binding) reversed impaired hindlimb perfusion and arteriogenesis induced by endothelial Raptor knockout in high-fat diet fed mice. CONCLUSION Collectively, our study demonstrated the crucial role of endothelial Raptor in promoting ischaemia-induced arteriogenesis in T2DM by mediating VEGFR2 signalling. Thus, endothelial Raptor is a novel therapeutic target for promoting arteriogenesis and ameliorating perfusion in T2DM.
Collapse
Affiliation(s)
- Ting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Jiachen Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Fangyuan Chang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Mengyu Sun
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jinlong He
- Department of Physiology and Pathophysiology, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| | - Ding Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Qixiangtai Rd 22nd, Tianjin 300070, China
| |
Collapse
|
3
|
Palzkill VR, Tan J, Yang Q, Morcos J, Laitano O, Ryan TE. Deletion of the aryl hydrocarbon receptor in endothelial cells improves ischemic angiogenesis in chronic kidney disease. Am J Physiol Heart Circ Physiol 2024; 326:H44-H60. [PMID: 37921663 PMCID: PMC11213484 DOI: 10.1152/ajpheart.00530.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Chronic kidney disease (CKD) is a strong risk factor for peripheral artery disease (PAD) that is associated with worsened clinical outcomes. CKD leads to the accumulation of tryptophan metabolites that are associated with adverse limb events in PAD and are ligands of the aryl hydrocarbon receptor (AHR), which may regulate ischemic angiogenesis. To test if endothelial cell-specific deletion of the AHR (AHRecKO) alters ischemic angiogenesis and limb function in mice with CKD subjected to femoral artery ligation. Male AHRecKO mice with CKD displayed better limb perfusion recovery and enhanced ischemic angiogenesis compared with wild-type mice with CKD. However, the improved limb perfusion did not result in better muscle performance. In contrast to male mice, deletion of the AHR in female mice with CKD had no impact on perfusion recovery or angiogenesis. With the use of primary endothelial cells from male and female mice, treatment with indoxyl sulfate uncovered sex-dependent differences in AHR activating potential and RNA sequencing revealed wide-ranging sex differences in angiogenic signaling pathways. Endothelium-specific deletion of the AHR improved ischemic angiogenesis in male, but not female, mice with CKD. There are sex-dependent differences in Ahr activating potential within endothelial cells that are independent of sex hormones.NEW & NOTEWORTHY This study provides novel insights into the mechanisms by which chronic kidney disease worsens ischemic limb outcomes in an experimental model of peripheral artery disease. Deletion of the aryl hydrocarbon receptor (AHR) in the endothelium improved ischemic angiogenesis suggesting that AHR inhibition could be a viable therapeutic target; however, this effect was only observed in male mice. Subsequent analysis in primary endothelial cells reveals sex differences in Ahr activating potential independent of sex hormones.
Collapse
Affiliation(s)
- Victoria R Palzkill
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Juliana Morcos
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
- Center for Exercise Science, University of Florida, Gainesville, Florida, United States
- The Myology Institute, University of Florida, Gainesville, Florida, United States
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, United States
- Center for Exercise Science, University of Florida, Gainesville, Florida, United States
- The Myology Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
4
|
Arderiu G, Civit-Urgell A, Badimon L. Adipose-Derived Stem Cells to Treat Ischemic Diseases: The Case of Peripheral Artery Disease. Int J Mol Sci 2023; 24:16752. [PMID: 38069074 PMCID: PMC10706341 DOI: 10.3390/ijms242316752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Critical limb ischemia incidence and prevalence have increased over the years. However, there are no successful treatments to improve quality of life and to reduce the risk of cardiovascular and limb events in these patients. Advanced regenerative therapies have focused their interest on the generation of new blood vessels to repair tissue damage through the use of stem cells. One of the most promising sources of stem cells with high potential in cell-based therapy is adipose-derived stem cells (ASCs). ASCs are adult mesenchymal stem cells that are relatively abundant and ubiquitous and are characterized by a multilineage capacity and low immunogenicity. The proangiogenic benefits of ASCs may be ascribed to: (a) paracrine secretion of proangiogenic molecules that may stimulate angiogenesis; (b) secretion of microvesicles/exosomes that are also considered as a novel therapeutic prospect for treating ischemic diseases; and (c) their differentiation capability toward endothelial cells (ECs). Although we know the proangiogenic effects of ASCs, the therapeutic efficacy of ASCs after transplantation in peripheral artery diseases patients is still relatively low. In this review, we evidence the potential therapeutic use of ASCs in ischemic regenerative medicine. We also highlight the main challenges in the differentiation of these cells into functional ECs. However, significant efforts are still needed to ascertain relevant transcription factors, intracellular signaling and interlinking pathways in endothelial differentiation.
Collapse
Affiliation(s)
- Gemma Arderiu
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| | - Anna Civit-Urgell
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Facultat de Medicina i Ciències de la Salut—Campus Clínic, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Lina Badimon
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Kastora SL, Eley J, Gannon M, Melvin R, Munro E, Makris SA. What Went Wrong with VEGF-A in Peripheral Arterial Disease? A Systematic Review and Biological Insights on Future Therapeutics. J Vasc Res 2022; 59:381-393. [PMID: 36380643 PMCID: PMC9808638 DOI: 10.1159/000527079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Of the 200 million patients worldwide affected by peripheral arterial disease (PAD), 4% will inevitably require major limb amputation. Previous systematic reviews presented a conflicting body of evidence in terms of vascular endothelial growth factor (VEGF) family member effects upon PAD natural progression. Despite that, modulation of intrinsic angiogenesis mechanisms targeting the VEGF family members still confers an attractive therapeutic target. The aim of the present study was to evaluate current evidence of VEGF modulation in the context of PAD. METHODS This is a systematic literature review conducted according to the PRISMA guidelines and registered under PROSPERO database [CRD42021285988]. Independent literature search was performed up to April 1, 2022, on six databases. A total of 22 eligible studies were identified [N: 3, interventional patient studies; N: 19, animal studies]. Animal studies were appraised by the SYRCLE risk of bias tool, while human participant studies were assessed by the Newcastle Ottawa scale. Overall, quality of evidence was deemed fair for both animal and human studies. Main study outcomes were percentage change of injured vessel lumen stenosis and neointimal area formation upon VEGF modulation (inhibition or activation) in comparison with control group. FINDINGS Nineteen animal models and three human participant studies were included in the systematic review and assessed separately. Positive modulation of VEGF-A in animal models resulted in a median decrease of 65.58% [95% CI 45.2; 71.87] in lumen stenosis [14 studies]. Furthermore, positive modulation of VEGF-A was found to reduce neointimal area proliferation by a median decrease of 63.41% [95% CI 41.6; 79.59] [14 studies]. Median end of study duration was 28 days [range: 14-84 days]. Data were insufficient to assess these outcomes with respect to VEGF-B or VEGF-C modulation. The limited number of available human studies presented inadequate outcome assessment despite their overall fair NOS grading. INTERPRETATION VEGF-A-positive modulation decreases lumen stenosis and neointimal hyperplasia in PAD simulation animal models. Previously identified variability among outcomes was found to strongly stem from the variability of experimental designs. Clinical applicability and safety profile of VEGF-A in the context of PAD remain to be defined by a robust and uniformly designed body of further animal model-based experiments.
Collapse
Affiliation(s)
- Stavroula L. Kastora
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK,*Stavroula L. Kastora,
| | - Jonathan Eley
- Department of General Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Martin Gannon
- Department of Vascular Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Ross Melvin
- Department of Vascular Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Euan Munro
- Department of Vascular Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | | |
Collapse
|
6
|
Angiogenesis in diabetic mouse model with critical limb ischemia; cell and gene therapy. Microvasc Res 2022; 141:104339. [DOI: 10.1016/j.mvr.2022.104339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/23/2022] [Accepted: 02/07/2022] [Indexed: 01/13/2023]
|
7
|
Bian D, Wu Y, Song G, Azizi R, Zamani A. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res Ther 2022; 13:24. [PMID: 35073970 PMCID: PMC8785459 DOI: 10.1186/s13287-021-02697-9] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, mesenchymal stromal cells (MSCs) and also their exosome has become a game-changing tool in the context of tissue engineering and regenerative medicine. MSCs due to their competencies to establish skin cells, such as fibroblast and keratinocyte, and also their unique attribute to suppress inflammation in wound site has attracted increasing attention among scholars. In addition, MSC's other capabilities to induce angiogenesis as a result of secretion of pro-angiogenic factors accompanied with marked anti-fibrotic activities, which mainly mediated by the releases matrix metalloproteinase (MMPs), make them a rational and effective strategy to accelerate wound healing with a small scar. Since the chief healing properties of the MSCs depend on their paracrine effects, it appears that MSCs-derived exosomes also can be an alternative option to support wound healing and skin regeneration as an innovative cell-free approach. Such exosomes convey functional cargos (e.g., growth factor, cytokine, miRNA, etc.) from MSCs to target cells, thereby affecting the recipient skin cells' biological events, such as migration, proliferation, and also secretion of ECM components (e.g., collagen). The main superiorities of exosome therapy over parental MSCs are the diminished risk of tumor formation and also lower immunogenicity. Herein, we deliver an overview of recent in vivo reports rendering the therapeutic benefits of the MSCs-based therapies to ease skin wound healing, and so improving quality of life among patients suffering from such conditions.
Collapse
Affiliation(s)
- Donghui Bian
- Department of Burns and Plastic Surgery, 960 Hospital of the People’s Liberation Army, Jinan, 250031 China
| | - Yan Wu
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013 China
| | - Guodong Song
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013 China
| | - Ramyar Azizi
- Department of Immunology, Medicine Faculty, Tabriz University of Medical Science, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Assis A, Camargo S, Margalit R, Mitrani E. Creation of a vascular inducing device using mesenchymal stem cells to induce angiogenesis. J Biosci Bioeng 2021; 132:408-416. [PMID: 34326013 DOI: 10.1016/j.jbiosc.2021.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Conventional treatments of peripheral vascular disease and coronary artery disease have partial success but are still limited. Methods to deliver angiogenic factors into ischemic areas using gene, protein and cell therapies are faced with difficult issues such a delivery, effective concentration and duration of action. Tissue engineering offers the possibility of creating a functional self-contained three-dimensional (3D) unit that works as a coordinated biological pump that can secrete a whole range of angiogenic factors. We report a tissue engineering approach using decellularized micro-fragments and mesenchymal stem cells (MSCs) to create a vascular inducing device (VID). Proteomic analysis of the decellularized micro-fragments and of the VIDs reveals a large number of extracellular-matrix (ECM) proteins. Moreover, the VIDs were found to transcribe and secrete a whole repertoire of angiogenic factors in a sustained manner. Furthermore, preliminary results of implantation VIDs into non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice indicate formation of vascular network at the site within a week. We propose that those VIDs could serve as a safe, localized, simple and powerful method for the treatment of certain types of vascular diseases.
Collapse
Affiliation(s)
- Assaf Assis
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel
| | - Sandra Camargo
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel
| | | | - Eduardo Mitrani
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Givat Ram Campus, Jerusalem 91904, Israel.
| |
Collapse
|
9
|
Qin F, Zhang W, Zhang M, Long X, Si L, Li Z, Huang J, Wang X. Adipose-Derived Stem Cells Improve the Aging Skin of Nude Mice by Promoting Angiogenesis and Reducing Local Tissue Water. Aesthet Surg J 2021; 41:NP905-NP913. [PMID: 33428732 DOI: 10.1093/asj/sjab001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) are considered promising cells for skin rejuvenation. However, whether the angiogenetic effect of ASCs plays an important role in the treatment of aging skin and its influence on skin tissue remain elusive. OBJECTIVES The aim of this study was to evaluate the effect of ASCs on angiogenesis and local tissue water (LTW) in the aging skin of nude mice. METHODS Twelve nude mice were randomly divided into a UVB-induced photoaging group and a natural aging group. After the mouse model had been established, ASCs and phosphate-buffered saline (PBS) were then each injected into different sides of the dorsal skin of the mice. Blood perfusion and LTW content were measured. After 7 weeks, mice were killed, and skin samples were collected to measure the thickness of the dermis, the density of the capillaries, and the expression of angiogenic growth factors. RESULTS ASC therapy significantly increased the thickness of the dermis, the number of capillaries, and the expression of some angiogenic growth factors (vascular endothelial growth factor, insulin-like growth factor 1, and epidermal growth factor). At 7 weeks after injection, blood perfusion was significantly higher on the side injected with ASCs than on the side injected with PBS. LTW content was increased in the PBS-injected side, but the ASC-injected side showed no significant changes over time. CONCLUSIONS ASCs increased dermal thickness, promoted angiogenesis, and reduced LTW content in the skin of photoaging mice, providing a potential clinical therapy for skin rejuvenation.
Collapse
Affiliation(s)
- Feng Qin
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Wenchao Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Mingzi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Loubin Si
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Zhenjiang Li
- Department of Vascular Surgery, First Affiliated Hospital of the Medical School of Zhejiang University, Hangzhou, China
| | - Jiuzuo Huang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xiaojun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Nammian P, Asadi-Yousefabad SL, Daneshi S, Sheikhha MH, Tabei SMB, Razban V. Comparative analysis of mouse bone marrow and adipose tissue mesenchymal stem cells for critical limb ischemia cell therapy. Stem Cell Res Ther 2021; 12:58. [PMID: 33436054 PMCID: PMC7805174 DOI: 10.1186/s13287-020-02110-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. METHODS For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. RESULTS Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. CONCLUSIONS Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.
Collapse
Affiliation(s)
- Pegah Nammian
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sajad Daneshi
- Postdoctoral Researcher, Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hasan Sheikhha
- Biotechnology Research Center, International Campus, Shahid Sadoughi University of MedicalSciences, Yazd, Iran
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Bagher Tabei
- Department of Genetics, Shiraz University of Medical Science, Shiraz, Iran.
- Maternal-fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Stem Cells Technology Research center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Abstract
Nanotechnology could offer a new complementary strategy for the treatment of vascular diseases including coronary, carotid, or peripheral arterial disease due to narrowing or blockage of the artery caused by atherosclerosis. These arterial diseases manifest correspondingly as angina and myocardial infarction, stroke, and intermittent claudication of leg muscles during exercise. The pathogenesis of atherosclerosis involves biological events at the cellular and molecular level, thus targeting these using nanomaterials precisely and effectively could result in a better outcome. Nanotechnology can mitigate the pathological events by enhancing the therapeutic efficacy of the therapeutic agent by delivering it at the point of a lesion in a controlled and efficacious manner. Further, combining therapeutics with imaging will enhance the theranostic ability in atherosclerosis. Additionally, nanoparticles can provide a range of delivery systems for genes, proteins, cells, and drugs, which individually or in combination can address various problems within the arteries. Imaging studies combined with nanoparticles helps in evaluating the disease progression as well as the response to the treatment because imaging and diagnostic agents can be delivered precisely to the targeted destinations via nanocarriers. This review focuses on the use of nanotechnology in theranostics of coronary artery and peripheral arterial disease.
Collapse
|
12
|
Hypothesizing the therapeutic potential of mesenchymal stem cells in oral submucous fibrosis. Med Hypotheses 2020; 144:110204. [PMID: 33254511 DOI: 10.1016/j.mehy.2020.110204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/31/2022]
Abstract
Oral submucous fibrosis is the direct consequence of a sustained pro-inflammatory environment characterized by excessive collagen deposition causing tissue fibrosis, and progressive degeneration of vital structures including muscle. The pathogenesis of oral submucous fibrosis is largely mediated by the pro-inflammatory, pro-fibrotic cytokines, excessive oxidative stress, abnormal angiogenesis, and epithelial to mesenchymal transition. Mesenchymal stem cells largely known for their regenerative potential have shown to have an immunomodulatory, anti-fibrotic, anti-oxidative, and angiogenic potential. Thus, mesenchymal stem cells, when introduced in an oral submucous fibrosis micro-environment, could potentially counter the progressive fibrosis. The present hypotheses discuss the various pathogenic aspects of oral submucous fibrosis and the properties of mesenchymal stem cells which could aid in halting the disease progression.
Collapse
|
13
|
Czapla J, Cichoń T, Pilny E, Jarosz-Biej M, Matuszczak S, Drzyzga A, Krakowczyk Ł, Smolarczyk R. Adipose tissue-derived stromal cells stimulated macrophages-endothelial cells interactions promote effective ischemic muscle neovascularization. Eur J Pharmacol 2020; 883:173354. [PMID: 32663541 DOI: 10.1016/j.ejphar.2020.173354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/31/2022]
Abstract
Neovascularization, the process of new blood vessels formation in response to hypoxia induced signals, is an essential step during wound healing or ischemia repair. It follows as a cascade of consecutive events leading to new blood vessels formation and their subsequent remodeling to a mature and functional state, enabling tissue regeneration. Any disruption in consecutive stages of neovascularization can lead to chronic wounds or impairment of tissue repair. In the study we try to explain the biological basis of accelerated blood vessels formation in ischemic tissue after adipose tissue-derived stromal cells (ADSCs) administration. Experiments were performed on mouse models of hindlimb ischemia. We have evaluated the level of immune cells (neutrophils, macrophages) infiltration. The novelty of our work was the assessment of bone marrow-derived stem/progenitor cells (BMDCs) infiltration and their contribution to the neovascularization process in ischemic tissue. We have noticed that ADSCs regulated immune response and affected the kinetics and ratio of macrophages population infiltrating ischemic tissue. Our research revealed that ADSCs promoted changes in the morphology of infiltrating macrophages and their tight association with forming blood vessels. We assume that recruited macrophages may take over the role of pericytes and stabilize the new blood vessel or even differentiate into endothelial cells, which in consequence can accelerate vascular formation upon ADSCs administration. Our findings indicate that administration of ADSCs into ischemic muscle influence spatio-temporal distribution of infiltrating cells (macrophages, neutrophils and BMDCs), which are involved in each step of vascular formation, promoting effective ischemic tissue neovascularization.
Collapse
Affiliation(s)
- Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland.
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland; Department of Organic Chemistry, Biochemistry and Biotechnology, Silesian University of Technology, Księdza Marcina Strzody 9 Street, 44-100, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Alina Drzyzga
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Łukasz Krakowczyk
- Department of Oncologic and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| |
Collapse
|
14
|
Bis(maltolato)oxovanadium(IV) Induces Angiogenesis via Phosphorylation of VEGFR2. Int J Mol Sci 2020; 21:ijms21134643. [PMID: 32629855 PMCID: PMC7370103 DOI: 10.3390/ijms21134643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 11/17/2022] Open
Abstract
VEGFR2 and VEGF-A play a pivotal role in the process of angiogenesis. VEGFR2 activation is regulated by protein tyrosine phosphatases (PTPs), enzymes that dephosphorylate the receptor and reduce angiogenesis. We aim to study the effect of PTPs blockade using bis(maltolato)oxovanadium(IV) (BMOV) on in vivo wound healing and in vitro angiogenesis. BMOV significantly improves in vivo wound closure by 45% in C57BL/6JRj mice. We found that upon VEGFR2 phosphorylation induced by endogenously produced VEGF-A, the addition of BMOV results in increased cell migration (45%), proliferation (40%) and tube formation (27%) in HUVECs compared to control. In a mouse ex vivo, aortic ring assay BMOV increased the number of sprouts by 3 folds when compared to control. However, BMOV coadministered with exogenous VEGF-A increased ECs migration, proliferation and tube formation by only 41%, 18% and 12% respectively and aortic ring sprouting by only 1-fold. We also found that BMOV enhances VEGFR2 Y951 and p38MAPK phosphorylation, but not ERK1/2. The level of phosphorylation of these residues was the same in the groups treated with BMOV supplemented with exogenous VEGF-A and exogenous VEGF-A only. Our study demonstrates that BMOV is able to enhance wound closure in vivo. Moreover, in the presence of endogenous VEGF-A, BMOV is able to stimulate in vitro angiogenesis by increasing the phosphorylation of VEGFR2 and its downstream proangiogenic enzymes. Importantly, BMOV had a stronger proangiogenic effect compared to its effect in coadministration with exogenous VEGF-A.
Collapse
|
15
|
Proust R, Ponsen AC, Rouffiac V, Schenowitz C, Montespan F, Ser-Le Roux K, De Leeuw F, Laplace-Builhé C, Mauduit P, Carosella ED, Banzet S, Lataillade JJ, Rouas-Freiss N, Uzan G, Peltzer J. Cord blood-endothelial colony forming cells are immunotolerated and participate at post-ischemic angiogenesis in an original dorsal chamber immunocompetent mouse model. Stem Cell Res Ther 2020; 11:172. [PMID: 32381102 PMCID: PMC7206734 DOI: 10.1186/s13287-020-01687-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/30/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cardiovascular diseases are the main cause of morbidity and mortality worldwide. Restoring blood supply to ischemic tissues is an essential goal for the successful treatment of these diseases. Growth factor or gene therapy efficacy remains controversial, but stem cell transplantation is emerging as an interesting approach to stimulate angiogenesis. Among the different stem cell populations, cord blood-endothelial progenitor cells (CB-EPCs) and more particularly cord blood-endothelial progenitor cell-derived endothelial colony forming cells (CB-ECFCs) have a great proliferative potential without exhibiting signs of senescence. Even if it was already described that CB-ECFCs were able to restore blood perfusion in hind-limb ischemia in an immunodeficient mouse model, until now, the immunogenic potential of allogenic CB-ECFCs remains controversial. Therefore, our objectives were to evaluate the immune tolerance potency of CB-ECFCs and their capacity to restore a functional vascular network under ischemic condition in immunocompetent mice. METHODS In vitro, the expression and secretion of immunoregulatory markers (HLA-G, IL-10, and TGF-β1) were evaluated on CB-ECFCs. Moreover, CB-ECFCs were co-cultured with activated peripheral blood mononuclear cells (PBMCs) for 6 days. PBMC proliferation was evaluated by [3H]-thymidine incorporation on the last 18 h. In vivo, CB-ECFCs were administered in the spleen and muscle of immunocompetent mice. Tissues were collected at day 14 after surgery. Finally, CB-ECFCs were injected intradermally in C57BL/6JRj mice close to ischemic macrovessel induced by thermal cauterization. Mice recovered until day 5 and were imaged, twice a week until day 30. RESULTS Firstly, we demonstrated that CB-ECFCs expressed HLA-G, IL-10, and TGF-β1 and secreted IL-10 and TGF-β1 and that they could display immunosuppressive properties in vitro. Secondly, we showed that CB-ECFCs could be tolerated until 14 days in immunocompetent mice. Thirdly, we revealed in an original ischemic model of dorsal chamber that CB-ECFCs were integrated in a new functional vascular network. CONCLUSION These results open up new perspectives about using CB-ECFCs as an allogeneic cell therapy product and gives new impulse to the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Richard Proust
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Anne-Charlotte Ponsen
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Valérie Rouffiac
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Chantal Schenowitz
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Florent Montespan
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Karine Ser-Le Roux
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Frédéric De Leeuw
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Corinne Laplace-Builhé
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Philippe Mauduit
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Edgardo D Carosella
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Sébastien Banzet
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Jean-Jacques Lataillade
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Juliette Peltzer
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France.
| |
Collapse
|
16
|
Circulating Metabolites Originating from Gut Microbiota Control Endothelial Cell Function. Molecules 2019; 24:molecules24213992. [PMID: 31694161 PMCID: PMC6864778 DOI: 10.3390/molecules24213992] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/15/2019] [Accepted: 11/03/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular functionality strictly depends on endothelial cell trophism and proper biochemical function. Any condition (environmental, pharmacological/toxicological, physical, or neuro-humoral) that changes the vascular endothelium has great consequences for the organism’s wellness and on the outcome and evolution of severe cardiovascular pathologies. Thus, knowledge of the mechanisms, both endogenous and external, that affect endothelial dysfunction is pivotal to preventing and treating these disorders. In recent decades, significant attention has been focused on gut microbiota and how these symbiotic microorganisms can influence host health and disease development. Indeed, dysbiosis has been reported to be at the base of a range of different pathologies, including pathologies of the cardiovascular system. The study of the mechanism underlying this relationship has led to the identification of a series of metabolites (released by gut bacteria) that exert different effects on all the components of the vascular system, and in particular on endothelial cells. The imbalance of factors promoting or blunting endothelial cell viability and function and angiogenesis seems to be a potential target for the development of new therapeutic interventions. This review highlights the circulating factors identified to date, either directly produced by gut microbes or resulting from the metabolism of diet derivatives as polyphenols.
Collapse
|
17
|
Yu Z, Witman N, Wang W, Li D, Yan B, Deng M, Wang X, Wang H, Zhou G, Liu W, Sahara M, Cao Y, Fritsche-Danielson R, Zhang W, Fu W, Chien KR. Cell-mediated delivery of VEGF modified mRNA enhances blood vessel regeneration and ameliorates murine critical limb ischemia. J Control Release 2019; 310:103-114. [PMID: 31425721 DOI: 10.1016/j.jconrel.2019.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/05/2019] [Accepted: 08/15/2019] [Indexed: 01/20/2023]
Abstract
Synthetic chemically modified mRNAs (modRNA) encoding vascular endothelial growth factor (VEGF) represents an alternative to gene therapy for the treatment of ischemic cardiovascular injuries. However, novel delivery approaches of modRNA are needed to improve therapeutic efficacy in the diseased setting. We hypothesized that cell-mediated modRNA delivery may enhance the in vivo expression kinetics of VEGF protein thus promoting more potent angiogenic effects. Here, we employed skin fibroblasts as a "proof of concept" to probe the therapeutic potential of a cell-mediated mRNA delivery system in a murine model of critical limb ischemia (CLI). We show that fibroblasts pre-treated with VEGF modRNA have the potential to fully salvage ischemic limbs. Using detailed molecular analysis we reveal that a fibroblast-VEGF modRNA combinatorial treatment significantly reduced tissue necrosis and dramatically improved vascular densities in CLI-injured limbs when compared to control and vehicle groups. Furthermore, fibroblast-delivered VEGF modRNA treatment increased the presence of Pax7+ satellite cells, indicating a possible correlation between VEGF and satellite cell activity. Our study is the first to demonstrate that a cell-mediated modRNA therapy could be an alternative advanced strategy for cardiovascular diseases.
Collapse
Affiliation(s)
- Ziyou Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Nevin Witman
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Dong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Bingqian Yan
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China; Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Mingwu Deng
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Xiangsheng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China; Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | - Regina Fritsche-Danielson
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Gothenburg, 43183, Sweden
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai 200011, China.
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China; Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China.
| | - Kenneth R Chien
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Johnson TK, Zhao L, Zhu D, Wang Y, Xiao Y, Oguljahan B, Zhao X, Kirlin WG, Yin L, Chilian WM, Liu D. Exosomes derived from induced vascular progenitor cells promote angiogenesis in vitro and in an in vivo rat hindlimb ischemia model. Am J Physiol Heart Circ Physiol 2019; 317:H765-H776. [PMID: 31418583 DOI: 10.1152/ajpheart.00247.2019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Induced vascular progenitor cells (iVPCs) were created as an ideal cell type for regenerative medicine and have been reported to positively promote collateral blood flow and improve cardiac function in a rat model of myocardial ischemia. Exosomes have emerged as a novel biomedicine that mimics the function of the donor cells. We investigated the angiogenic activity of exosomes from iPVCs (iVPC-Exo) as a cell-free therapeutic approach for ischemia. Exosomes from iVPCs and rat aortic endothelial cells (RAECs) were isolated using a combination of ultrafiltration and size-exclusion chromatography. Nanoparticle tracking analysis revealed that exosome isolates fell within the exosomal diameter (<150 nm). These exosomes contained known markers Alix and TSG101, and their morphology was validated using transmission electron microscopy. When compared with RAECs, iVPCs significantly increased the secretion of exosomes. Cardiac microvascular endothelial cells and aortic ring explants were pretreated with RAEC-Exo or iVPC-Exo, and basal medium was used as a control. iVPC-Exo exerted an in vitro angiogenic effect on the proliferation, tube formation, and migration of endothelial cells and stimulated microvessel sprouting in an ex vivo aortic ring assay. Additionally, iVPC-Exo increased blood perfusion in a hindlimb ischemia model. Proangiogenic proteins (pentraxin-3 and insulin-like growth factor-binding protein-3) and microRNAs (-143-3p, -291b, and -20b-5p) were found to be enriched in iVPC-Exo, which may mediate iVPC-Exo induced vascular growth. Our findings demonstrate that treatment with iVPC-Exo promotes angiogenesis in vitro, ex vivo, and in vivo. Collectively, these findings indicate a novel cell-free approach for therapeutic angiogenesis.NEW & NOTEWORTHY The results of this work demonstrate exosomes as a novel physiological mechanism by which induced vascular progenitor cells exert their angiogenic effect. Moreover, angiogenic cargo of proteins and microRNAs may define the biological contributors in activating endothelial cells to form a new capillary plexus for ischemic vascular diseases.
Collapse
Affiliation(s)
- Takerra K Johnson
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Lina Zhao
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Dihan Zhu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Yang Wang
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Yan Xiao
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Babayewa Oguljahan
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Ward G Kirlin
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Dong Liu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia.,Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
19
|
Lin Q, Wang DG, Zhang ZQ, Liu DP. Applications of Virus Vector-Mediated Gene Therapy in China. Hum Gene Ther 2019; 29:98-109. [PMID: 29284296 DOI: 10.1089/hum.2017.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to the increased safety and efficiency of virus vectors, virus vector-mediated gene therapy is now widely used for various diseases, including monogenic diseases, complex disorders, and infectious diseases. Recent gene therapy trials have shown significant therapeutic benefits, and Chinese researchers have contributed significantly to this progress. This review highlights disease applications and strategies for virus vector-mediated gene therapy in preclinical studies and clinical trials in China.
Collapse
Affiliation(s)
- Qiong Lin
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deng-Gao Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhu-Qin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Zhang J, Zhang H, Chen Y, Fu J, Lei Y, Sun J, Tang B. Platelet‑derived growth factor D promotes the angiogenic capacity of endothelial progenitor cells. Mol Med Rep 2018; 19:125-132. [PMID: 30483778 PMCID: PMC6297765 DOI: 10.3892/mmr.2018.9692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Neovascularization and re-endothelialization rely on endothelial progenitor cells (EPCs). However, the recruitment and angiogenic roles of EPCs are subject to regulation through the vascular microenvironment, which remains largely unknown. Platelet-derived growth factor D (PDGF-D) is a new member of the PDGF family that binds the PDGFR-β homodimer. However, it remains unknown whether and how it affects the angiogenic capacity of EPCs and participates in tube-like formation. EPCs were derived from rat bone marrow cells, and the gain-of-function approach was used to study the effects of PDGF-D on the biological activities of EPCs. EPCs that stably express PDGF-D were generated by lentiviral-mediated transduction, and the expression levels were evaluated by western blotting and reverse transcription, followed by real-time quantitative polymerase chain reaction (RT-qPCR). The biological activities of EPCs evaluated in the present study included proliferation, adhesion, migration, tube formation and senescence. Furthermore, the downstream signaling of PDGF-D was explored by western blot analysis. The results revealed that the lentiviral-mediated expression of PDGF-D in the microenvironment promoted the migration, proliferation, adhesion and tube formation of EPCs. In addition, PDGF-D suppressed the senescence of EPCs. Mechanistically, PDGF-D induced the phosphorylation of several signaling molecules, including STAT3, AKT, ERK1/2, mTOR and GSK-3β, suggesting that PDGF-D enhanced the angiogenic function of EPCs through PDGF receptor-dependent and -independent signaling pathways. In conclusion, PDGF-D promotes the angiogenic capacity of EPCs, including proliferation, migration, adhesion and tube formation, and thereby contributes to angiogenesis.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Haolong Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yikuan Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jian Fu
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yu Lei
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianming Sun
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Bo Tang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
21
|
Liu J, Wada Y, Katsura M, Tozawa H, Erwin N, Kapron CM, Bao G, Liu J. Rho-Associated Coiled-Coil Kinase (ROCK) in Molecular Regulation of Angiogenesis. Am J Cancer Res 2018; 8:6053-6069. [PMID: 30613282 PMCID: PMC6299434 DOI: 10.7150/thno.30305] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Identified as a major downstream effector of the small GTPase RhoA, Rho-associated coiled-coil kinase (ROCK) is a versatile regulator of multiple cellular processes. Angiogenesis, the process of generating new capillaries from the pre-existing ones, is required for the development of various diseases such as cancer, diabetes and rheumatoid arthritis. Recently, ROCK has attracted attention for its crucial role in angiogenesis, making it a promising target for new therapeutic approaches. In this review, we summarize recent advances in understanding the role of ROCK signaling in regulating the permeability, migration, proliferation and tubulogenesis of endothelial cells (ECs), as well as its functions in non-ECs which constitute the pro-angiogenic microenvironment. The therapeutic potential of ROCK inhibitors in angiogenesis-related diseases is also discussed.
Collapse
|
22
|
Fierro FA, Magner N, Beegle J, Dahlenburg H, Logan White J, Zhou P, Pepper K, Fury B, Coleal-Bergum DP, Bauer G, Gruenloh W, Annett G, Pifer C, Nolta JA. Mesenchymal stem/stromal cells genetically engineered to produce vascular endothelial growth factor for revascularization in wound healing and ischemic conditions. Transfusion 2018; 59:893-897. [PMID: 30383901 DOI: 10.1111/trf.14914] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) may be able to improve ischemic conditions as they can actively seek out areas of low oxygen and secrete proangiogenic factors. In more severe trauma and chronic cases, however, cells alone may not be enough. Therefore, we have combined the stem cell and angiogenic factor approaches to make a more potent therapy. We developed an engineered stem cell therapy product designed to treat critical limb ischemia that could also be used in trauma-induced scarring and fibrosis where additional collateral blood flow is needed following damage to and blockage of the primary vessels. We used MSCs from normal human donor marrow and engineered them to produce high levels of the angiogenic factor vascular endothelial growth factor (VEGF). The MSC/VEGF product has been successfully developed and characterized using good manufacturing practice (GMP)-compliant methods, and we have completed experiments showing that MSC/VEGF significantly increased blood flow in the ischemic limb of immune deficient mice, compared to the saline controls in each study. We also performed safety studies demonstrating that the injected product does not cause harm and that the cells remain around the injection site for more than 1 month after hypoxic preconditioning. An on-demand formulation system for delivery of the product to clinical sites that lack cell processing facilities is in development.
Collapse
Affiliation(s)
- Fernando A Fierro
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Nataly Magner
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Julie Beegle
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Heather Dahlenburg
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Jeannine Logan White
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Ping Zhou
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Karen Pepper
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Brian Fury
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | | | - Gerhard Bauer
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - William Gruenloh
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Geralyn Annett
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Christy Pifer
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| | - Jan A Nolta
- Institute for Regenerative Cures, University of California at Davis, Sacramento, California
| |
Collapse
|
23
|
Use of Nutraceuticals in Angiogenesis-Dependent Disorders. Molecules 2018; 23:molecules23102676. [PMID: 30340320 PMCID: PMC6222874 DOI: 10.3390/molecules23102676] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
The term of angiogenesis refers to the growth of new vessels from pre-existing capillaries. The phenomenon is necessary for physiological growth, repair and functioning of our organs. When occurring in a not regulated manner, it concurs to pathological conditions as tumors, eye diseases, chronic degenerative disorders. On the contrary insufficient neovascularization or endothelial disfunction accompanies ischemic and metabolic disorders. In both the cases an inflammatory and oxidative condition exists in supporting angiogenesis deregulation and endothelial dysfunction. The use of nutraceuticals with antioxidant and anti-inflammatory activities can be a therapeutic option to maintain an adequate vascularization and endothelial cell proper functioning or to blunt aberrant angiogenesis. A revision of the updated literature reports on nutraceuticals to guide endothelial cell wellness and to restore physiological tissue vascularization is the objective of this paper. The critical aspects as well as lacking data for human use will be explored from a pharmacological perspective.
Collapse
|
24
|
Wang LS, Wang H, Zhang QL, Yang ZJ, Kong FX, Wu CT. Hepatocyte Growth Factor Gene Therapy for Ischemic Diseases. Hum Gene Ther 2018; 29:413-423. [DOI: 10.1089/hum.2017.217] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Li-Sheng Wang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- School of Nursing, Jilin University, Jilin, P.R. China
| | - Hua Wang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Qing-Lin Zhang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Zhi-Jian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Fan-Xuan Kong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Chu-Tse Wu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| |
Collapse
|
25
|
Wang C, Li Y, Yang M, Zou Y, Liu H, Liang Z, Yin Y, Niu G, Yan Z, Zhang B. Efficient Differentiation of Bone Marrow Mesenchymal Stem Cells into Endothelial Cells in Vitro. Eur J Vasc Endovasc Surg 2017; 55:257-265. [PMID: 29208350 DOI: 10.1016/j.ejvs.2017.10.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/18/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Endothelial cells (ECs) play an important role in neovascularisation, but are too limited in number for adequate therapeutic applications. Mesenchymal stem cells (MSCs) have the potential to differentiate into endothelial lineage cells, which makes them attractive candidates for therapeutic angiogenesis. The aim of this study was to investigate efficient differentiation of MSCs into ECs by inducing medium in vitro. METHODS MSCs were isolated from bone marrow by density gradient centrifugation. The characterisation of the MSCs was determined by their cluster of differentiation (CD) marker profile. Inducing medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin like growth factor (IGF), epidermal growth factor (EGF), ascorbic acid, and heparin was applied to differentiate the MSCs into ECs. Endothelial differentiation was quantitatively evaluated using flow cytometry. Real time quantitative PCR (qRT-PCR) was used to analyse mRNA expression of endothelial markers. Tube formation assay was further performed to examine the functional status of the differentiated MSCs. RESULTS Flow cytometry analysis demonstrated that CD31+ and CD34+ cells increased steadily from 12% at 3 days, to 40% at 7 days, and to 60% at 14 days. Immunofluorescence staining further confirmed the expression of CD31 and CD34. qRT-PCR showed that expression of von Willebrand factor (vWF), vascular endothelial cadherin (VE-cadherin) and vascular endothelial growth factor receptor-2 (VEGFR-2) were significantly higher in the induced MSCs group compared with the uninduced MSCs group. The functional behavior of the differentiated cells was tested by tube formation assay in vitro on matrigel. Induced MSCs were capable of developing capillary networks, and progressive formation of vessel like structures was associated with increased EC population. CONCLUSIONS These results provide a method to efficiently promote differentiation of MSCs into ECs in vitro for potential application in the treatment of peripheral arterial disease.
Collapse
Affiliation(s)
- Chengen Wang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Yuan Li
- Department of Haematology, Peking University First Hospital, Beijing, China.
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Yinghua Zou
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China.
| | - Huihui Liu
- Department of Haematology, Peking University First Hospital, Beijing, China
| | - Zeyin Liang
- Department of Haematology, Peking University First Hospital, Beijing, China
| | - Yue Yin
- Department of Haematology, Peking University First Hospital, Beijing, China
| | - Guochen Niu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Ziguang Yan
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
26
|
Zahavi-Goldstein E, Blumenfeld M, Fuchs-Telem D, Pinzur L, Rubin S, Aberman Z, Sher N, Ofir R. Placenta-derived PLX-PAD mesenchymal-like stromal cells are efficacious in rescuing blood flow in hind limb ischemia mouse model by a dose- and site-dependent mechanism of action. Cytotherapy 2017; 19:1438-1446. [PMID: 29122516 DOI: 10.1016/j.jcyt.2017.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND In peripheral artery disease (PAD), blockage of the blood supply to the limbs, most frequently the legs, leads to impaired blood flow and tissue ischemia. Pluristem's PLX-PAD cells are placenta-derived mesenchymal stromal-like cells currently in clinical trials for the treatment of peripheral artery diseases. METHODS In this work, the hind limb ischemia (HLI) mouse model was utilized to study the efficacy and mechanism of action of PLX-PAD cells. ELISA assays were performed to characterize and quantitate PLX-PAD secretions in vitro. RESULTS PLX-PAD cells administered intramuscularly rescued blood flow to the lower limb after HLI induction in a dose-dependent manner. While rescue of blood flow was site-dependent, numerous administration regimes enabled rescue of blood flow, indicating a systemic effect mediated by PLX-PAD secretions. Live PLX-PAD cells were more efficacious than cell lysate in rescuing blood flow, indicating the importance of prolonged cytokine secretion for maximal blood flow recovery. In vitro studies showed a multifactorial secretion profile including numerous pro-angiogenic proteins; these are likely involved in the PLX-PAD mechanism of action. DISCUSSION Live PLX-PAD cells were efficacious in rescuing blood flow after the induction of HLI in the mouse model in a dose- and site-dependent manner. The fact that various administration routes of PLX-PAD rescued blood flow indicates that the mechanism of action likely involves one of systemic secretions which promote angiogenesis. Taken together, the data support the further clinical testing of PLX-PAD cells for PAD indications.
Collapse
|
27
|
Anderson EM, Silva EA, Hao Y, Martinick KD, Lewin SA, Stafford AG, Doherty EG, Wang L, Doherty EJ, Grossman PM, Mooney DJ. VEGF and IGF Delivered from Alginate Hydrogels Promote Stable Perfusion Recovery in Ischemic Hind Limbs of Aged Mice and Young Rabbits. J Vasc Res 2017; 54:288-298. [PMID: 28930755 PMCID: PMC5642984 DOI: 10.1159/000479869] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
Biomaterial-based delivery of angiogenic growth factors restores perfusion more effectively than bolus delivery methods in rodent models of peripheral vascular disease, but the same success has not yet been demonstrated in clinically relevant studies of aged or large animals. These studies explore, in clinically relevant models, a therapeutic angiogenesis strategy for the treatment of peripheral vascular disease that overcomes the challenges encountered in previous clinical trials. Alginate hydrogels providing sustained release of vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF) were injected into ischemic hind limbs in middle-aged and old mice, and also in young rabbits, as a test of the scalability of this local growth factor treatment. Spontaneous perfusion recovery diminished with increasing age, and only the combination of VEGF and IGF delivery from gels significantly rescued perfusion in middle-aged (13 months) and old (20 months) mice. In rabbits, the delivery of VEGF alone or in combination with IGF from alginate hydrogels, at a dose 2 orders of magnitude lower than the typical doses used in past rabbit studies, enhanced perfusion recovery when given immediately after surgery, or as a treatment for chronic ischemia. Capillary density measurements and angiographic analysis demonstrated the benefit of gel delivery. These data together suggest that alginate hydrogels providing local delivery of low doses of VEGF and IGF constitute a safe and effective treatment for hind-limb ischemia in clinically relevant animal models, thereby supporting the potential clinical translation of this concept.
Collapse
Affiliation(s)
- Erin M Anderson
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Eduardo A Silva
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Yibai Hao
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Kathleen D Martinick
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Sarah A Lewin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Alexander G Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Elisabeth G Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Lin Wang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Edward J Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Paul M Grossman
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Dave J Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| |
Collapse
|
28
|
Rong SL, Wang XL, Zhang CY, Song ZH, Cui LH, He XF, Li XJ, Du HJ, Li B. Transplantation of HGF gene-engineered skeletal myoblasts improve infarction recovery in a rat myocardial ischemia model. PLoS One 2017; 12:e0175807. [PMID: 28459804 PMCID: PMC5411067 DOI: 10.1371/journal.pone.0175807] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Skeletal myoblast transplantation seems a promising approach for the repair of myocardial infarction (MI). However, the low engraftment efficacy and impaired angiogenic ability limit the clinical efficiency of the myoblasts. Gene engineering with angiogenic growth factors promotes angiogenesis and enhances engraftment of transplanted skeletal myoblasts, leading to improved infarction recovery in myocardial ischemia. The present study evaluated the therapeutic effects of hepatocyte growth factor (HGF) gene-engineered skeletal myoblasts on tissue regeneration and restoration of heart function in a rat MI model. METHODS AND RESULTS The skeletal myoblasts were isolated, expanded, and transduced with adenovirus carrying the HGF gene (Ad-HGF). Male SD rats underwent ligation of the left anterior descending coronary artery. After 2 weeks, the surviving rats were randomized into four groups and treated with skeletal myoblasts by direct injection into the myocardium. The survival and engraftment of skeletal myoblasts were determined by real-time PCR and in situ hybridization. The cardiac function with hemodynamic index and left ventricular architecture were monitored; The adenovirus-mediated-HGF gene transfection increases the HGF expression and promotes the proliferation of skeletal myoblasts in vitro. Transplantation of HGF-engineered skeletal myoblasts results in reduced infarct size and collagen deposition, increased vessel density, and improved cardiac function in a rat MI model. HGF gene modification also increases the myocardial levels of HGF, VEGF, and Bcl-2 and enhances the survival and engraftment of skeletal myoblasts. CONCLUSIONS HGF engineering improves the regenerative effect of skeletal myoblasts on MI by enhancing their survival and engraftment ability.
Collapse
Affiliation(s)
- Shu-Ling Rong
- Department of Cardiology, Heping Hospital and Institute of cardiovascular disease, Changzhi Medical College, Changzhi, China
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiao-Lin Wang
- Department of Pediatrics, Heping Hospital and Institute of cardiovascular disease, Changzhi Medical College, Changzhi, China
| | - Cui-Ying Zhang
- Department of Physiology, Changzhi Medical College, Changzhi, China
| | - Zhuo-Hui Song
- Department of Physiology, Changzhi Medical College, Changzhi, China
| | - Lu-Hua Cui
- Department of Cardiology, Heping Hospital and Institute of cardiovascular disease, Changzhi Medical College, Changzhi, China
| | - Xiao-Feng He
- Department of Research, Heping Hospital, Changzhi Medical College, Changzhi, China
| | - Xu-Jiong Li
- Department of Physiology, Changzhi Medical College, Changzhi, China
| | - Hui-Jin Du
- Department of Cardiology, Heping Hospital and Institute of cardiovascular disease, Changzhi Medical College, Changzhi, China
| | - Bao Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
29
|
Histopathological Analysis of Skeletal Muscle Biopsy of Patient with Peripheral Arterial Disease before and after Peripheral Blood Stem Cells Intramuscular Injection. BIONANOSCIENCE 2017. [DOI: 10.1007/s12668-016-0336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
Samura M, Hosoyama T, Takeuchi Y, Ueno K, Morikage N, Hamano K. Therapeutic strategies for cell-based neovascularization in critical limb ischemia. J Transl Med 2017; 15:49. [PMID: 28235425 PMCID: PMC5324309 DOI: 10.1186/s12967-017-1153-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
Critical limb ischemia (CLI) causes severe ischemic rest pain, ulcer, and gangrene in the lower limbs. In spite of angioplasty and surgery, CLI patients without suitable artery inflow or enough vascular bed in the lesions are often forced to undergo amputation of a major limb. Cell-based therapeutic angiogenesis has the potential to treat ischemic lesions by promoting the formation of collateral vessel networks and the vascular bed. Peripheral blood mononuclear cells and bone marrow-derived mononuclear cells are the most frequently employed cell types in CLI clinical trials. However, the clinical outcomes of cell-based therapeutic angiogenesis using these cells have not provided the promised benefits for CLI patients, reinforcing the need for novel cell-based therapeutic angiogenesis strategies to cure untreatable CLI patients. Recent studies have demonstrated the possible enhancement of therapeutic efficacy in ischemic diseases by preconditioned graft cells. Moreover, judging from past clinical trials, the identification of adequate transplant timing and responders to cell-based therapy is important for improving therapeutic outcomes in CLI patients in clinical settings. Thus, to establish cell-based therapeutic angiogenesis as one of the most promising therapeutic strategies for CLI patients, its advantages and limitations should be taken into account.
Collapse
Affiliation(s)
- Makoto Samura
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tohru Hosoyama
- Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan. .,Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan. .,Center for Regenerative Medicine, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Yuriko Takeuchi
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Noriyasu Morikage
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
31
|
Hsu PY, Hsi E, Wang TM, Lin RT, Liao YC, Juo SHH. MicroRNA let-7g possesses a therapeutic potential for peripheral artery disease. J Cell Mol Med 2016; 21:519-529. [PMID: 27696675 PMCID: PMC5323674 DOI: 10.1111/jcmm.12997] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/23/2016] [Indexed: 12/28/2022] Open
Abstract
Peripheral artery disease (PAD) is a manifestation of systemic atherosclerosis and conveys a significant health burden globally. Critical limb ischaemia encompasses the most severe consequence of PAD. Our previous studies indicate that microRNA let‐7g prevents atherosclerosis and improves endothelial functions. This study aimed to investigate whether and how let‐7g therapy may improve blood flow to ischaemic limbs. The present study shows that let‐7g has multiple pro‐angiogenic effects on mouse ischaemic limb model and could be a potential therapeutic agent for PAD. Mice receiving intramuscular injection of let‐7g had more neovascularization, better local perfusion and increased recruitment of endothelial progenitor cells after hindlimb ischaemia. The therapeutic effects of let‐7g's on angiogenesis are mediated by multiple regulatory machinery. First, let‐7g increased expression of vascular endothelial growth factor‐A (VEGF‐A) and VEGF receptor‐2 (VEGFR‐2) through targeting their upstream regulators HIF‐3α and TP53. In addition, let‐7g affected the splicing factor SC35 which subsequently enhanced the alternative splicing of VEGF‐A from the anti‐angiogenic isoform VEGF‐A165b towards the pro‐angiogenic isoform VEGF‐A164a. The pleiotropic effects of let‐7g on angiogenesis imply that let‐7g may possess a therapeutic potential in ischaemic diseases.
Collapse
Affiliation(s)
- Po-Yuan Hsu
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Edward Hsi
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ming Wang
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Ruey-Tay Lin
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chu Liao
- Department of Neurology, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Suh-Hang H Juo
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
32
|
Pereira ARS, Mendes TF, Ministro A, Teixeira M, Filipe M, Santos JM, Bárcia RN, Goyri-O'Neill J, Pinto F, Cruz PE, Cruz HJ, Santos SCR. Therapeutic angiogenesis induced by human umbilical cord tissue-derived mesenchymal stromal cells in a murine model of hindlimb ischemia. Stem Cell Res Ther 2016; 7:145. [PMID: 27680210 PMCID: PMC5041588 DOI: 10.1186/s13287-016-0410-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/09/2016] [Indexed: 11/10/2022] Open
Abstract
Background Mesenchymal stem cells derived from human umbilical cord tissue, termed UCX®, have the potential to promote a full range of events leading to tissue regeneration and homeostasis. The main goal of this work was to investigate UCX® action in experimentally induced hindlimb ischemia (HLI). Methods UCX®, obtained by using a proprietary technology developed by ECBio (Amadora, Portugal), were delivered via intramuscular injection to C57BL/6 females after unilateral HLI induction. Perfusion recovery, capillary and collateral density increase were evaluated by laser doppler, CD31 immunohistochemistry and diaphonisation, respectively. The activation state of endothelial cells (ECs) was analysed after EC isolation by laser capture microdissection microscopy followed by RNA extraction, cDNA synthesis and quantitative RT-PCR analysis. The UCX®-conditioned medium was analysed on Gallios flow cytometer. The capacity of UCX® in promoting tubulogenesis and EC migration was assessed by matrigel tubule formation and wound-healing assay, respectively. Results We demonstrated that UCX® enhance angiogenesis in vitro via a paracrine effect. Importantly, after HLI induction, UCX® improve blood perfusion by stimulating angiogenesis and arteriogenesis. This is achieved through a new mechanism in which durable and simultaneous upregulation of transforming growth factor β2, angiopoietin 2, fibroblast growth factor 2, and hepatocyte growth factor, in endothelial cells is induced by UCX®. Conclusions In conclusion, our data demonstrate that UCX® improve the angiogenic potency of endothelial cells in the murine ischemic limb suggesting the potential of UCX® as a new therapeutic tool for critical limb ischemia.
Collapse
Affiliation(s)
- Ana Rita S Pereira
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Teresa F Mendes
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Augusto Ministro
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.,Centro Hospitalar Lisboa Norte, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal
| | - Mariana Teixeira
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Mariana Filipe
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Jorge M Santos
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Rita N Bárcia
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - J Goyri-O'Neill
- Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Fausto Pinto
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.,Faculdade de Medicina da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Pedro E Cruz
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Helder J Cruz
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal. .,Faculdade de Medicina da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.
| |
Collapse
|
33
|
Sessions JW, Lewis TE, Skousen CS, Hope S, Jensen BD. The effect of injection speed and serial injection on propidium iodide entry into cultured HeLa and primary neonatal fibroblast cells using lance array nanoinjection. SPRINGERPLUS 2016; 5:1093. [PMID: 27468394 PMCID: PMC4947087 DOI: 10.1186/s40064-016-2757-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/05/2016] [Indexed: 01/01/2023]
Abstract
Background Although site-directed genetic engineering has greatly improved in recent years, particularly with the implementation of CRISPR-Cas9, the ability to deliver these molecular constructs to a wide variety of cell types without adverse reaction is still a challenge. One non-viral transfection method designed to address this challenge is a MEMS based biotechnology described previously as lance array nanoinjection (LAN). LAN delivery of molecular loads is based upon the combinational use of electrical manipulation of loads of interest and physical penetration of target cell membranes. This work explores an original procedural element to nanoinjection by investigating the effects of the speed of injection and also the ability to serially inject the same sample. Results Initial LAN experimentation demonstrated that injecting at speeds of 0.08 mm/s resulted in 99.3 % of cultured HeLa 229 cells remaining adherent to the glass slide substrate used to stage the injection process. These results were then utilized to examine whether or not target cells could be injected multiple times (1, 2, and 3 times) since the injection process was not pulling the cells off of the glass slide. Using two different current control settings (1.5 and 3.0 mA) and two different cell types (HeLa 229 cells and primary neonatal fibroblasts [BJ(ATCC® CRL-2522™)], treatment samples were injected with propidium iodide (PI), a cell membrane impermeable nucleic acid dye, to assess the degree of molecular load delivery. Results from the serial injection work indicate that HeLa cells treated with 3.0 mA and injected twice (×2) had the greatest mean PI uptake of 60.47 % and that neonatal fibroblasts treated with the same protocol reached mean PI uptake rates of 20.97 %. Conclusions Both experimental findings are particularly useful because it shows that greater molecular modification rates can be achieved by multiple, serial injections via a slower injection process.
Collapse
Affiliation(s)
- John W Sessions
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 USA
| | - Tyler E Lewis
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 USA
| | - Craig S Skousen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602 USA
| | - Sandra Hope
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602 USA
| | - Brian D Jensen
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 USA
| |
Collapse
|
34
|
Lee JC, Cha CI, Kim D, Choe SY. Therapeutic effects of umbilical cord blood derived mesenchymal stem cell-conditioned medium on pulmonary arterial hypertension in rats. J ANAT SOC INDIA 2016. [DOI: 10.1016/j.jasi.2016.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Flex A, Biscetti F, Iachininoto MG, Nuzzolo ER, Orlando N, Capodimonti S, Angelini F, Valentini CG, Bianchi M, Larocca LM, Martini M, Teofili L. Human cord blood endothelial progenitors promote post-ischemic angiogenesis in immunocompetent mouse model. Thromb Res 2016; 141:106-11. [PMID: 26994683 DOI: 10.1016/j.thromres.2016.03.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Human cord blood (CB) endothelial colony forming cells (ECFCs) are endowed with high vascular regenerative ability in immunodeficient mice, but their immunogenicity and susceptibility to rejection in immunocompetent models has yet to be explored. METHODS We injected CB ECFCs in non-immuno-suppressed C57BL/6J mice after having induced the hindlimb ischemia and we investigated their contribution to the recovery from the ischemic injury. Human ECFCs (hECFCs) were administered by intramuscular injection and hindlimb blood perfusion was measured by laser Doppler analysis at 7-day intervals for 28days after treatment. Mice were sacrificed after 7 and 28days and immunohistochemistry for specific human (CD31) and mouse (von Willebrand factor) endothelial antigens was carried out. Before euthanasia, blood samples to assess cytokines and angiogenic growth factor levels were collected. RESULTS Mice injected with hECFCs showed a prompter and greater recovery of blood flow than controls. Several endothelial cells of human origin were detected at day7 after injection and their number declined progressively. Likewise, a progressive increase of mouse-derived vascular structures were observed, paralleled by the amplified endogenous production of various soluble mediators of angiogenesis, including Vascular Endothelial Growth Factor and Fibroblast Growth Factor. CONCLUSIONS Overall, our findings are consistent with the hypothesis that human ECFCs might expand the endogenous vascular repair potential of recipients and support their possible HLA-independent unconventional use.
Collapse
Affiliation(s)
- Andrea Flex
- Department of Internal Medicine, Catholic University, Rome, Italy
| | | | | | | | | | | | - Flavia Angelini
- Department of Internal Medicine, Catholic University, Rome, Italy
| | | | - Maria Bianchi
- Institute of Hematology, Catholic University, Rome, Italy
| | | | | | | |
Collapse
|
36
|
Tao H, Han Z, Han ZC, Li Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int 2016; 2016:1314709. [PMID: 26880933 PMCID: PMC4736816 DOI: 10.1155/2016/1314709] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/04/2015] [Accepted: 11/29/2015] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown their therapeutic potency for treatment of cardiovascular diseases owing to their low immunogenicity, ease of isolation and expansion, and multipotency. As multipotent progenitors, MSCs have revealed their ability to differentiate into various cell types and could promote endogenous angiogenesis via microenvironmental modulation. Studies on cardiovascular diseases have demonstrated that transplanted MSCs could engraft at the injured sites and differentiate into cardiomyocytes and endothelial cells as well. Accordingly, several clinical trials using MSCs have been performed and revealed that MSCs may improve relevant clinical parameters in patients with vascular diseases. To fully comprehend the characteristics of MSCs, understanding their intrinsic property and associated modulations in tuning their behaviors as well as functions is indispensable for future clinical translation of MSC therapy. This review will focus on recent progresses on endothelial differentiation and potential clinical application of MSCs, with emphasis on therapeutic angiogenesis for treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyan Tao
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| | - Zhibo Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhong Chao Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| |
Collapse
|
37
|
Abstract
In 2015, it can be said that the diabetic foot is no longer the Cinderella of diabetic complications. Thirty years ago there was little evidence-based research taking place on the diabetic foot, and there were no international meetings addressing this topic. Since then, the biennial Malvern Diabetic Foot meetings started in 1986, the American Diabetes Association founded their Foot Council in 1987, and the European Association for the Study of Diabetes established a Foot Study Group in 1998. The first International Symposium on the Diabetic Foot in The Netherlands was convened in 1991, and this was soon followed by the establishment of the International Working Group on the Diabetic Foot that has produced useful guidelines in several areas of investigation and the management of diabetic foot problems. There has been an exponential rise in publications on diabetic foot problems in high impact factor journals, and a comprehensive evidence-base now exists for many areas of treatment. Despite the extensive evidence available, it, unfortunately, remains difficult to demonstrate that most types of education are efficient in reducing the incidence of foot ulcers. However, there is evidence that education as part of a multi-disciplinary approach to diabetic foot ulceration plays a pivotal role in incidence reduction. With respect to treatment, strong evidence exists that offloading is the best modality for healing plantar neuropathic foot ulcers, and there is also evidence from two randomized controlled trials to support the use of negative-pressure wound therapy in complex post-surgical diabetic foot wounds. Hyperbaric oxygen therapy exhibits the same evidence level and strength of recommendation. International guidelines exist on the management of infection in the diabetic foot. Many randomized trials have been performed, and these have shown that the agents studied generally produced comparable results, with the exception of one study in which tigecycline was shown to be clinically inferior to ertapenem ± vancomycin. Similarly, there are numerous types of wound dressings that might be used in treatment and which have shown efficacy, but no single type (or brand) has shown superiority over others. Peripheral artery disease is another major contributory factor in the development of ulceration, and its presence is a strong predictor of non-healing and amputation. Despite the proliferation of endovascular procedures in addition to open revascularization, many patients continue to suffer from severely impaired perfusion and exhaust all treatment options. Finally, the question of the true aetiopathogenesis of Charcot neuroarthropathy remains enigmatic, although much work is currently being undertaken in this area. In this area, it is most important to remember that a clinically uninfected, warm, insensate foot in a diabetic patient should be considered as a Charcot foot until proven otherwise, and, as such, treated with offloading, preferably in a cast.
Collapse
Affiliation(s)
- K Markakis
- Manchester Royal Infirmary, Manchester, UK
| | - F L Bowling
- Manchester Royal Infirmary, Manchester, UK
- University of Manchester, Manchester, UK
| | - A J M Boulton
- Manchester Royal Infirmary, Manchester, UK
- University of Manchester, Manchester, UK
| |
Collapse
|
38
|
Hepatocyte Growth Factor Effects on Mesenchymal Stem Cells Derived from Human Arteries: A Novel Strategy to Accelerate Vascular Ulcer Wound Healing. Stem Cells Int 2015; 2016:3232859. [PMID: 26788066 PMCID: PMC4691635 DOI: 10.1155/2016/3232859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/11/2015] [Accepted: 08/23/2015] [Indexed: 12/25/2022] Open
Abstract
Vascular ulcers are a serious complication of peripheral vascular disease, especially in diabetics. Several approaches to treat the wounds are proposed but they show poor outcomes and require long healing times. Hepatocyte Growth Factor/Scatter Factor (HGF/SF) is a pleiotropic cytokine exerting many biological activities through the c-Met receptor. This study was aimed at verifying whether HGF/SF influences proliferation, migration, and angiogenesis on mesenchymal stem cells isolated from human arteries (hVW-MSCs). hVW-MSCs were exposed to NIBSC HGF/SF (2.5, 5, 10, and 70 ng/mL) from 6 hrs to 7 days. HGF and c-MET mRNA and protein expression, cell proliferation (Alamar Blue and Ki-67 assay), migration (scratch and transwell assays), and angiogenesis (Matrigel) were investigated. hVW-MSCs displayed stemness features and expressed HGF and c-MET. HGF/SF did not increase hVW-MSC proliferation, whereas it enhanced the cell migration, the formation of capillary-like structures, and the expression of angiogenic markers (vWF, CD31, and KDR). The HGF/SF effects on hVW-MSC migration and angiogenic potential are of great interest to accelerate wound healing process. Local delivery of HGF/SF could therefore improve the healing of unresponsive vascular ulcers.
Collapse
|
39
|
Cell Therapy in Patients with Critical Limb Ischemia. Stem Cells Int 2015; 2015:931420. [PMID: 26300924 PMCID: PMC4537766 DOI: 10.1155/2015/931420] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/30/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022] Open
Abstract
Critical limb ischemia (CLI) represents the most advanced stage of peripheral arterial obstructive disease (PAOD) with a severe obstruction of the arteries which markedly reduces blood flow to the extremities and has progressed to the point of severe rest pain and/or even tissue loss. Recent therapeutic strategies have focused on restoring this balance in favor of tissue survival using exogenous molecular and cellular agents to promote regeneration of the vasculature. These are based on stimulation of angiogenesis by extracellular and cellular components. This review article carries out a systematic analysis of the most recent scientific literature on the application of stem cells in patients with CLI. The results obtained from the detailed analysis of the recent literature data have confirmed the beneficial role of cell therapy in reducing the rate of major amputations in patients with CLI and improving their quality of life.
Collapse
|
40
|
Kolluru GK, Bir SC, Yuan S, Shen X, Pardue S, Wang R, Kevil CG. Cystathionine γ-lyase regulates arteriogenesis through NO-dependent monocyte recruitment. Cardiovasc Res 2015; 107:590-600. [PMID: 26194202 DOI: 10.1093/cvr/cvv198] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/02/2015] [Indexed: 12/29/2022] Open
Abstract
AIMS Hydrogen sulfide (H2S) is a vasoactive gasotransmitter that is endogenously produced in the vasculature by the enzyme cystathionine γ-lyase (CSE). However, the importance of CSE activity and local H2S generation for ischaemic vascular remodelling remains completely unknown. In this study, we examine the hypothesis that CSE critically regulates ischaemic vascular remodelling involving H2S-dependent mononuclear cell regulation of arteriogenesis. METHODS AND RESULTS Arteriogenesis including mature vessel density, collateral formation, blood flow, and SPY angiographic blush rate were determined in wild-type (WT) and CSE knockout (KO) mice at different time points following femoral artery ligation (FAL). The role of endogenous H2S in regulation of IL-16 expression and subsequent recruitment of monocytes, and expression of VEGF and bFGF in ischaemic tissues, were determined along with endothelial progenitor cell (CD34/Flk1) formation and function. FAL of WT mice significantly increased CSE activity, expression and endogenous H2S generation in ischaemic tissues, and monocyte infiltration, which was absent in CSE-deficient mice. Treatment of CSE KO mice with the polysulfide donor diallyl trisulfide restored ischaemic vascular remodelling, monocyte infiltration, and cytokine expression. Importantly, exogenous H2S therapy restored nitric oxide (NO) bioavailability in CSE KO mice that was responsible for monocyte recruitment and arteriogenesis. CONCLUSION Endogenous CSE/H2S regulates ischaemic vascular remodelling mediated during hind limb ischaemia through NO-dependent monocyte recruitment and cytokine induction revealing a previously unknown mechanism of arteriogenesis.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, LSU Health Sciences Center Shreveport Center for Cardiovascular Diseases and Sciences, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, USA
| | - Shyamal C Bir
- Department of Pathology, LSU Health Sciences Center Shreveport
| | - Shuai Yuan
- Department of Pathology, LSU Health Sciences Center Shreveport Center for Cardiovascular Diseases and Sciences, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, USA
| | - Xinggui Shen
- Department of Pathology, LSU Health Sciences Center Shreveport
| | - Sibile Pardue
- Department of Pathology, LSU Health Sciences Center Shreveport
| | - Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, ON, Canada
| | - Christopher G Kevil
- Department of Pathology, LSU Health Sciences Center Shreveport Center for Cardiovascular Diseases and Sciences, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, USA
| |
Collapse
|
41
|
Prokosch V, Stupp T, Spaniol K, Pham E, Nikol S. Angiogenic gene therapy does not cause retinal pathology. J Gene Med 2015; 16:309-16. [PMID: 25322754 DOI: 10.1002/jgm.2806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/22/2014] [Accepted: 08/26/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The potential negative influence of angiogenic gene therapy on the development or progression of retinal pathologies such as diabetic retinopathy (DR) or age-related macular degeneration (AMD) has led to the systematic exclusion of affected patients from trials. We investigated the role of nonviral fibroblast factor 1 (NV1FGF) in two phase II, multinational, double-blind, randomized, placebo-controlled, gene therapy trials (TALISMAN 201 and 211). METHODS One hundred and fifty-two subjects with critical limb ischemia or claudication were randomized to receive eight intramuscular injections of 2.5 ml of NV1FGF at 0.2 mg/ml or 0.4 mg/dl or placebo. One hundred and fifty-two patients received a plasmid dose of NV1FGF of up to 32 mg or placebo. All patients underwent a systematic ophthalmologic examination at baseline and at 3, 6 or 12 months following gene therapy. Twenty-six of these patients (Münster subgroup) received a retinal fluorescence angiography at baseline and at final examination. RESULTS Among those 26 patients, four of nine patients with diabetes suffered from nonproliferative DR. Three patients showed non-exsudative AMD. No change of retinal morphology or function was observed in Münster subgroup of both TALISMAN trials independent of the intramuscular NV1FGF dosage applied. CONCLUSIONS Angiogenic gene therapy using NV1FGF is safe even in diabetics.
Collapse
|
42
|
A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int J Mol Sci 2015; 16:11294-322. [PMID: 25993296 PMCID: PMC4463701 DOI: 10.3390/ijms160511294] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 03/29/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
Peripheral artery disease (PAD) is due to the blockage of the arteries supplying blood to the lower limbs usually secondary to atherosclerosis. The most severe clinical manifestation of PAD is critical limb ischemia (CLI), which is associated with a risk of limb loss and mortality due to cardiovascular events. Currently CLI is mainly treated by surgical or endovascular revascularization, with few other treatments in routine clinical practice. There are a number of problems with current PAD management strategies, such as the difficulty in selecting the appropriate treatments for individual patients. Many patients undergo repeated attempts at revascularization surgery, but ultimately require an amputation. There is great interest in developing new methods to identify patients who are unlikely to benefit from revascularization and to improve management of patients unsuitable for surgery. Circulating biomarkers that predict the progression of PAD and the response to therapies could assist in the management of patients. This review provides an overview of the pathophysiology of PAD and examines the association between circulating biomarkers and PAD presence, severity and prognosis. While some currently identified circulating markers show promise, further larger studies focused on the clinical value of the biomarkers over existing risk predictors are needed.
Collapse
|
43
|
Abstract
Vascular endothelial growth factor A (VEGF-A) is a potent proangiogenic cytokine elevated in patients with peripheral artery disease (PAD). A new study links impaired vascular regrowth in PAD to increased expression of an antiangiogenic splice variant of VEGF-A.
Collapse
|
44
|
Sanada F, Taniyama Y, Kanbara Y, Otsu R, Ikeda-Iwabu Y, Carracedo M, Rakugi H, Morishita R. Gene therapy in peripheral artery disease. Expert Opin Biol Ther 2015; 15:381-90. [PMID: 25633211 DOI: 10.1517/14712598.2015.1007039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Despite the remarkable progress of medicine and endovascular procedures for revascularization, patients with critical limb ischemia (CLI) remain at high risk for amputation and often have a low quality of life due to pain and ulcers in the ischemic leg. Thus, a novel strategy for generating new blood vessels in CLI patients without treatment options is vital. Pre-clinical studies and Phase I clinical trials using VEGF and fibroblast growth factor (FGF) demonstrated promising results; however, more rigorous Phase II and III clinical trials failed to demonstrate benefits for CLI patients. Recently, two multicenter, double-blind, placebo-controlled clinical trials in Japan (Phase III) and the USA (Phase II) showed the benefits of hepatocyte growth factor (HGF) gene therapy for CLI patients. Although the number of patients included in these trials was relatively small, these results imply a distinct beneficial function for HGF over other angiogenic growth factors in a clinical setting. AREAS COVERED In this review, data from Phase I-III clinical trials of gene therapy for patients with peripheral artery disease (PAD) are examined. In addition, the potential mechanisms behind the success or failure of clinical trials are discussed. EXPERT OPINION Compared with VEGF and FGF, HGF has a unique molecular effect on inflammation, fibrosis and cell senescence under pathological conditions. These features may explain the clinical benefits of HGF in PAD patients.
Collapse
Affiliation(s)
- Fumihiro Sanada
- Osaka University Graduate School of Medicine, Department of Clinical Gene Therapy , Suita, Osaka 565-0871 , Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Maijub JG, Boyd NL, Dale JR, Hoying JB, Morris ME, Williams SK. Concentration-Dependent Vascularization of Adipose Stromal Vascular Fraction Cells. Cell Transplant 2014; 24:2029-39. [PMID: 25397993 DOI: 10.3727/096368914x685401] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adipose-derived stromal vascular fraction (SVF) cells have been shown to self-associate to form vascular structures under both in vitro and in vivo conditions. The angiogenic (new vessels from existing vessels) and vasculogenic (new vessels through self-assembly) potential of the SVF cell population may provide a cell source for directly treating (i.e., point of care without further cell isolation) ischemic tissues. However the correct dosage of adipose SVF cells required to achieve a functional vasculature has not been established. Accordingly, in vitro and in vivo dose response assays were performed evaluating the SVF cell vasculogenic potential. Serial dilutions of freshly isolated rat adipose SVF cells were plated on growth factor reduced Matrigel and vasculogenesis, assessed as cellular tube-like network assembly, was quantified after 3 days of culture. This in vitro vasculogenesis assay indicated that rat SVF cells reached maximum network length at a concentration of 2.5 × 10(5) cells/ml and network maintained at the higher concentrations tested. The same concentrations of rat and human SVF cells were used to evaluate vasculogenesis in vivo. SVF cells were incorporated into collagen gels and subcutaneously implanted into Rag1 immunodeficient mice. The 3D confocal images of harvested constructs were evaluated to quantify dose dependency of SVF cell vasculogenesis potential. Rat- and human-derived SVF cells yielded a maximum vasculogenic potential at 1 × 10(6) and 4 × 10(6) cells/ml, respectively. No adverse reactions (e.g., toxicity, necrosis, tumor formation) were observed at any concentration tested. In conclusion, the vasculogenic potential of adipose-derived SVF cell populations is dose dependent.
Collapse
Affiliation(s)
- John G Maijub
- Cardiovascular Innovation Institute, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | | | | | | |
Collapse
|
46
|
Rümenapf G, Morbach S. What can I do with a patient with diabetes and critically impaired limb perfusion who cannot be revascularized? INT J LOW EXTR WOUND 2014; 13:378-89. [PMID: 25326447 DOI: 10.1177/1534734614554283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A patient with limb-threatening diabetic foot syndrome in whom relevant peripheral arterial occlusive disease is proven should receive arterial revascularization as soon as possible to avoid major amputation. However, 3 conditions may make it impossible to effectively restore limb perfusion: the patient will not profit from arterial revascularization owing to excessive tissue loss or sepsis ("too late"), the patient cannot be revascularized due to severe comorbidities ("too sick"), and arterial reconstruction is impossible because of technical and anatomical shortcomings in a patient who is otherwise fit for operation ("nonreconstructible limb perfusion"). This review outlines the therapeutic options specifically in the third group of patients in whom no technical option for surgical or endovascular revascularization exists.
Collapse
Affiliation(s)
- Gerhard Rümenapf
- Gefäßzentrum Oberrhein, Diakonissen-Stiftungs-Krankenhaus, Speyer, Germany
| | | |
Collapse
|
47
|
Abstract
The application of gene- and cell-based therapies to promote angiogenesis is a novel concept to treat lower-limb critical limb ischemia (CLI) and may provide an unmet need for patients with no options for revascularization. Proof of concept was demonstrated in animal models resulting in clinical trials that have confirmed the feasibility and short-term efficacy of intramuscular injection of angiogenetic tissue growth factors or bone marrow stem cells. The safety of these biologic therapies has been demonstrated in randomized clinical trials with no "off-target" angiogenesis, growth of occult tumors, or progression of diabetic retinopathy. Current phase III randomized clinical trials using a DNA plasmid with the hepatocyte growth factor gene or bone marrow aspirate concentrate of mesenchymal cells are designed to address several crucial issues, including proper patient selection criteria, relevant clinical endpoints, and long-term efficacy. Because effectiveness of these novel therapies remains to be established, ongoing and future randomized clinical trials should be placebo-controlled, investigator-blinded, and have amputation-free survival as the primary endpoint. Further development of efficient gene transfer techniques and keeping transplanted stem cells healthy have the potential to make biologic therapies more robust in promoting angiogenesis, tissue regeneration, and resolution of CLI symptoms. If sustained efficacy can be demonstrated, new therapeutic strategies for patients with CLI will be available for clinicians, ie, limb revascularization using angiogenic gene or stem cell therapy alone, or in conjunction with endovascular intervention.
Collapse
Affiliation(s)
- Sae Hee Ko
- Division of Vascular and Endovascular Surgery, University of California, San Diego, School of Medicine, Sulpizio Cardiovascular Center, 7404 Medical Center Drive, Mail Code 7403, La Jolla, CA 92037
| | - Dennis F Bandyk
- Division of Vascular and Endovascular Surgery, University of California, San Diego, School of Medicine, Sulpizio Cardiovascular Center, 7404 Medical Center Drive, Mail Code 7403, La Jolla, CA 92037.
| |
Collapse
|
48
|
Rask-Andersen M, Zhang J, Fabbro D, Schiöth HB. Advances in kinase targeting: current clinical use and clinical trials. Trends Pharmacol Sci 2014; 35:604-20. [PMID: 25312588 DOI: 10.1016/j.tips.2014.09.007] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 11/30/2022]
Abstract
Phosphotransferases, also known as kinases, are the most intensively studied protein drug target category in current pharmacological research, as evidenced by the vast number of kinase-targeting agents enrolled in active clinical trials. This development has emerged following the great success of small-molecule, orally available protein kinase inhibitors for the treatment of cancer, starting with the introduction of imatinib (Gleevec®) in 2003. The pharmacological utility of kinase-targeting has expanded to include treatment of inflammatory diseases, and rapid development is ongoing for kinase-targeted therapies in a broad array of indications in ophthalmology, analgesia, central nervous system (CNS) disorders, and the complications of diabetes, osteoporosis, and otology. In this review we highlight specifically the kinase drug targets and kinase-targeting agents being explored in current clinical trials. This analysis is based on a recent estimate of all established and clinical trial drug mechanisms of action, utilizing private and public databases to create an extensive dataset detailing aspects of more than 3000 approved and experimental drugs.
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, Biomedicinska Centrum (BMC), Uppsala 751 24, Sweden.
| | - Jin Zhang
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, Biomedicinska Centrum (BMC), Uppsala 751 24, Sweden; Department of Chemistry, Umeå Universitet, 901 87 Umeå, Sweden
| | - Doriano Fabbro
- PIQUR Therapeutics AG, Hohe Winde-Strasse 120, 4059 Basel, Switzerland
| | - Helgi B Schiöth
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, Biomedicinska Centrum (BMC), Uppsala 751 24, Sweden
| |
Collapse
|