1
|
Massidda MW, Demkov A, Sices A, Lee M, Lee J, Paull TT, Kim J, Baker AB. Mechanical Rejuvenation of Mesenchymal Stem Cells from Aged Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597781. [PMID: 38895474 PMCID: PMC11185588 DOI: 10.1101/2024.06.06.597781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mesenchymal stem cells (MSC) are an appealing therapeutic cell type for many diseases. However, patients with poor health or advanced age often have MSCs with poor regenerative properties. A major limiter of MSC therapies is cellular senescence, which is marked by limited proliferation capability, diminished multipotency, and reduced regenerative properties. In this work, we explored the ability of applied mechanical forces to reduce cellular senescence in MSCs. Our studies revealed that mechanical conditioning caused a lasting enhancement in proliferation, overall cell culture expansion potential, multipotency, and a reduction of senescence in MSCs from aged donors. Mechanistic studies suggested that these functional enhancements were mediated by oxidative stress and DNA damage repair signaling with mechanical load altering the expression of proteins of the sirtuin pathway, the DNA damage repair protein ATM, and antioxidant proteins. In addition, our results suggest a biophysical mechanism in which mechanical stretch leads to improved recognition of damaged DNA in the nucleus. Analysis of the cells through RNA-seq and ATAC-seq, demonstrated that mechanical loading alters the cell's genetic landscape to cause broad shifts in transcriptomic patterns that related to senescence. Overall, our results demonstrate that mechanical conditioning can rejuvenate mesenchymal stem cells derived from aged patients and improve their potential as a therapeutic cell type. GRAPHICAL ABSTRACT
Collapse
|
2
|
Friend NE, Beamish JA, Margolis EA, Schott NG, Stegemann JP, Putnam AJ. Pre-cultured, cell-encapsulating fibrin microbeads for the vascularization of ischemic tissues. J Biomed Mater Res A 2024; 112:549-561. [PMID: 37326361 PMCID: PMC10724379 DOI: 10.1002/jbm.a.37580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
There is a significant clinical need to develop effective vascularization strategies for tissue engineering and the treatment of ischemic pathologies. In patients afflicted with critical limb ischemia, comorbidities may limit common revascularization strategies. Cell-encapsulating modular microbeads possess a variety of advantageous properties, including the ability to support prevascularization in vitro while retaining the ability to be injected in a minimally invasive manner in vivo. Here, fibrin microbeads containing human umbilical vein endothelial cells (HUVEC) and bone marrow-derived mesenchymal stromal cells (MSC) were cultured in suspension for 3 days (D3 PC microbeads) before being implanted within intramuscular pockets in a SCID mouse model of hindlimb ischemia. By 14 days post-surgery, animals treated with D3 PC microbeads showed increased macroscopic reperfusion of ischemic foot pads and improved limb salvage compared to the cellular controls. Delivery of HUVEC and MSC via microbeads led to the formation of extensive microvascular networks throughout the implants. Engineered vessels of human origins showed evidence of inosculation with host vasculature, as indicated by erythrocytes present in hCD31+ vessels. Over time, the total number of human-derived vessels within the implant region decreased as networks remodeled and an increase in mature, pericyte-supported vascular structures was observed. Our findings highlight the potential therapeutic benefit of developing modular, prevascularized microbeads as a minimally invasive therapeutic for treating ischemic tissues.
Collapse
Affiliation(s)
- Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Jeffrey A. Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Emily A. Margolis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | | | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| |
Collapse
|
3
|
Zhang Y, Li X, Tian H, Xi M, Zhou J, Li H. p53 Activation Facilitates Transdifferentiation of Human Cardiac Fibroblasts into Endothelial Cells. Tissue Eng Part A 2024; 30:330-339. [PMID: 37819701 DOI: 10.1089/ten.tea.2023.0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Vascular endothelial cells (ECs), locating at the inner side of vascular lumen, play critical roles in maintaining vascular function and participate in tissue repair and neovascularization. Although increasing studies have shown positive effects of transplantation of vascular ECs or their precursor cells on neovascularization and functional recovery of ischemic tissues, the quantity of in vivo ECs is limited and their quality is affected by age, gender, disease, and others, which hinder their clinical application and further study. Chemical transdifferentiation is a promising approach to generate patient-specific cells. In this process, somatic cells are directly converted into desired cell types without the risk of tumorigenicity by pluripotent cell transplantation and exogenous gene introduction by transgene technology. In the present study, we derived ECs from human cardiac fibroblasts (CFs) through an optimized chemical induction method. The derived ECs expressed endothelial specific markers, took up low-density lipoprotein, secreted angiogenic cytokines under hypoxic condition, and formed microvessels in vitro and in vivo. This CF-EC transition bypassed pluripotency and germ layer differentiation, but underwent a stage of endothelialization. Although p53 maintained the same level during the period of CF-EC transdifferentiation, we could modulate p53 transcriptional activity to further improve cell transition efficiency, which mainly functioned at the later stage of endothelialization. Optimization and exploring the regulatory mechanism of CF-EC transition complement each other, which not only broadens the sources of patient-specific ECs but also provides valuable references for the in vivo direct transdifferentiation study and the elucidation of endothelial development and dysfunction. Impact statement This study provides an optimized chemical induction method to derive endothelial cells (ECs) from human cardiac fibroblasts (CFs), which not only broadens the sources of patient-specific ECs but also provides a good research model of mesenchymal-endothelial transition. Studying the molecular process and regulatory mechanism of CF-EC transdifferentiation will provide valuable references for the in vivo direct transdifferentiation for clinical therapy and deepen the understanding of endothelial development and dysfunction.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Histology and Embryology and School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Xuefeng Li
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Hong Tian
- Department of Histology and Embryology and School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Miaomiao Xi
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jinsong Zhou
- Department of Histology and Embryology and School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Hai Li
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
Huang NF, Stern B, Oropeza BP, Zaitseva TS, Paukshto MV, Zoldan J. Bioengineering Cell Therapy for Treatment of Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:e66-e81. [PMID: 38174560 PMCID: PMC10923024 DOI: 10.1161/atvbaha.123.318126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Peripheral artery disease is an atherosclerotic disease associated with limb ischemia that necessitates limb amputation in severe cases. Cell therapies comprised of adult mononuclear or stromal cells have been clinically tested and show moderate benefits. Bioengineering strategies can be applied to modify cell behavior and function in a controllable fashion. Using mechanically tunable or spatially controllable biomaterials, we highlight examples in which biomaterials can increase the survival and function of the transplanted cells to improve their revascularization efficacy in preclinical models. Biomaterials can be used in conjunction with soluble factors or genetic approaches to further modulate the behavior of transplanted cells and the locally implanted tissue environment in vivo. We critically assess the advances in bioengineering strategies such as 3-dimensional bioprinting and immunomodulatory biomaterials that can be applied to the treatment of peripheral artery disease and then discuss the current challenges and future directions in the implementation of bioengineering strategies.
Collapse
Affiliation(s)
- Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Brett Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | | | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| |
Collapse
|
5
|
Kizub IV. Induced pluripotent stem cells for cardiovascular therapeutics: Progress and perspectives. REGULATORY MECHANISMS IN BIOSYSTEMS 2023; 14:451-468. [DOI: 10.15421/10.15421/022366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The discovery of methods for reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) opens up prospects of developing personalized cell-based therapy options for a variety of human diseases as well as disease modeling and new drug discovery. Like embryonic stem cells, iPSCs can give rise to various cell types of the human body and are amenable to genetic correction. This allows usage of iPSCs in the development of modern therapies for many virtually incurable human diseases. The review summarizes progress in iPSC research in the context of application in the cardiovascular field including modeling cardiovascular disease, drug study, tissue engineering, and perspectives for personalized cardiovascular medicine.
Collapse
|
6
|
Zhang W, Huang X. Stem cell-based drug delivery strategy for skin regeneration and wound healing: potential clinical applications. Inflamm Regen 2023; 43:33. [PMID: 37391780 DOI: 10.1186/s41232-023-00287-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Stem cell-based therapy is widely accepted to be a promising strategy in tissue regenerative medicine. Nevertheless, there are several obstacles to applying stem cells in skin regeneration and wound healing, which includes determining the optimum source, the processing and administration methods of stem cells, and the survival and functions of stem cells in wound sites. Owing to the limitations of applying stem cells directly, this review aims to discuss several stem cell-based drug delivery strategies in skin regeneration and wound healing and their potential clinical applications. We introduced diverse types of stem cells and their roles in wound repair. Moreover, the stem cell-based drug delivery systems including stem cell membrane-coated nanoparticles, stem cell-derived extracellular vesicles, stem cell as drug carriers, scaffold-free stem cell sheets, and stem cell-laden scaffolds were further investigated in the field of skin regeneration and wound healing. More importantly, stem cell membrane-coating nanotechnology confers great advantages compared to other drug delivery systems in a broad field of biomedical contexts. Taken together, the stem cell-based drug delivery strategy holds great promise for treating skin regeneration and wound healing.
Collapse
Affiliation(s)
- Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Chan AH, Hu C, Chiang GC, Ekweume C, Huang NF. Chronic nicotine impairs the angiogenic capacity of human induced pluripotent stem cell-derived endothelial cells in a murine model of peripheral arterial disease. JVS Vasc Sci 2023; 4:100115. [PMID: 37519333 PMCID: PMC10372313 DOI: 10.1016/j.jvssci.2023.100115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/05/2023] [Indexed: 08/01/2023] Open
Abstract
Objective Lifestyle choices such as tobacco and e-cigarette use are a risk factor for peripheral arterial disease (PAD) and may influence therapeutic outcomes. The effect of chronic nicotine exposure on the angiogenic capacity of human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) was assessed in a murine model of PAD. Methods Mice were exposed to nicotine or phosphate-buffered saline (PBS) for 28 days, followed by induction of limb ischemia and iPSC-EC transplantation. Cells were injected into the ischemic limb immediately after induction of hindlimb ischemia and again 7 days later. Limb perfusion was assessed by laser Doppler spectroscopy, and transplant cell survival was monitored for 14 days afterward using bioluminescence imaging, followed by histological analysis of angiogenesis. Results Transplant cell retention progressively decreased over time after implantation based on bioluminescence imaging, and there were no significant differences in cell survival between mice with chronic exposure to nicotine or PBS. However, compared with mice without nicotine exposure, mice with prior nicotine exposure had had an impaired therapeutic response to iPSC-EC therapy based on decreased vascular perfusion recovery. Mice with nicotine exposure, followed by cell transplantation, had significantly lower mean perfusion ratio after 14 days (0.47 ± 0.07) compared with mice undergoing cell transplantation without prior nicotine exposure (0.79 ± 0.11). This finding was further supported by histological analysis of capillary density, in which animals with prior nicotine exposure had a lower capillary density (45.9 ± 4.7 per mm2) compared with mice without nicotine exposure (66.5 ± 8.1 per mm2). Importantly, the ischemic limbs mice exposed to nicotine without cell therapy also showed significant impairment in perfusion recovery after 14 days, compared with mice that received PBS + iPSC-EC treatment. This result suggested that mice without chronic nicotine exposure could respond to iPSC-EC implantation into the ischemic limb by inducing perfusion recovery, whereas mice with chronic nicotine exposure did not respond to iPSC-EC therapy. Conclusions Together, these findings show that chronic nicotine exposure adversely affects the ability of iPSC-EC therapy to promote vascular perfusion recovery and angiogenesis in a murine PAD model.
Collapse
Affiliation(s)
- Alex H.P. Chan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Caroline Hu
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Gladys C.F. Chiang
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Chisomaga Ekweume
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
- College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
- Department of Chemical Engineering, Stanford University, Stanford, CA
| |
Collapse
|
8
|
Jung C, Oh JE, Lee S, Yoon YS. Generation and Application of Directly Reprogrammed Endothelial Cells. Korean Circ J 2022; 52:643-658. [PMID: 36097834 PMCID: PMC9470489 DOI: 10.4070/kcj.2022.0190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/15/2022] Open
Abstract
Cell-based therapy has emerged as a promising option for treating advanced ischemic cardiovascular disease by inducing vascular regeneration. However, clinical trials with adult cells turned out disappointing in general. As a newer approach, direct reprogramming has emerged to efficiently generate endothelial cells (ECs), which can promote neovascularization and vascular regeneration. This review provides recent updates on the direct endothelial reprogramming. In general, directly reprogrammed ECs can be generated by two approaches: one by transitioning through a plastic intermediate state and the other in a one-step transition without any intermediate states toward pluripotency. Moreover, the methods to deliver reprogramming factors and chemicals for the fate conversion are highlighted. Next, the therapeutic effects of the directly reprogrammed ECs on animal models are reviewed in detail. Other applications using directly reprogrammed ECs, such as tissue engineering and disease modeling, are also discussed. Lastly, the remaining questions and foremost challenges are addressed.
Collapse
Affiliation(s)
- Cholomi Jung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Eun Oh
- Research and Development Center, KarisBio Inc., Seoul, Korea
| | - Sangho Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Research and Development Center, KarisBio Inc., Seoul, Korea
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
9
|
Enhancement strategy for effective vascular regeneration following myocardial infarction through a dual stem cell approach. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1165-1178. [PMID: 35974098 PMCID: PMC9440102 DOI: 10.1038/s12276-022-00827-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 11/08/2022]
Abstract
Since an impaired coronary blood supply following myocardial infarction (MI) negatively affects heart function, therapeutic neovascularization is considered one of the major therapeutic strategies for cell-based cardiac repair. Here, to more effectively achieve therapeutic neovascularization in ischemic hearts, we developed a dual stem cell approach for effective vascular regeneration by utilizing two distinct types of stem cells, CD31+-endothelial cells derived from human induced pluripotent stem cells (hiPSC-ECs) and engineered human mesenchymal stem cells that continuously secrete stromal derived factor-1α (SDF-eMSCs), to simultaneously promote natal vasculogenesis and angiogenesis, two core mechanisms of neovascularization. To induce more comprehensive vascular regeneration, we intramyocardially injected hiPSC-ECs to produce de novo vessels, possibly via vasculogenesis, and a 3D cardiac patch encapsulating SDF-eMSCs (SDF-eMSC-PA) to enhance angiogenesis through prolonged secretion of paracrine factors, including SDF-1α, was implanted into the epicardium of ischemic hearts. We verified that hiPSC-ECs directly contribute to de novo vessel formation in ischemic hearts, resulting in enhanced cardiac function. In addition, the concomitant implantation of SDF1α-eMSC-PAs substantially improved the survival, retention, and vasculogenic potential of hiPSC-ECs, ultimately achieving more comprehensive neovascularization in the MI hearts. Of note, the newly formed vessels through the dual stem cell approach were significantly larger and more functional than those formed by hiPSC-ECs alone. In conclusion, these results provide compelling evidence that our strategy for effective vascular regeneration can be an effective means to treat ischemic heart disease. A treatment involving two different types of stem cells leads to repairing failed hearts by making new functional blood vessels. Researchers at the City University of Hong Kong and the Catholic University of Korea induced heart attacks in rats before injecting the hearts with endothelial cells derived from human induced pluripotent stem cells, specialized to form blood vessels. These cells successfully induced the formation of new blood vessels in the damaged hearts. The researchers combined this treatment with a cardiac patch containing engineered human adult stem cells, which improved the survival and performance of the endothelial cells. And this dual stem cell treatment resulted in enhanced cardiac function and a higher number of larger and stronger new blood vessels than those produced by the single-cell treatment suggesting an effective way to repair failed hearts.
Collapse
|
10
|
Yu Y, Li X, Li Y, Wei R, Li H, Liu Z, Zhang Y. Derivation and Characterization of Endothelial Cells from Porcine Induced Pluripotent Stem Cells. Int J Mol Sci 2022; 23:ijms23137029. [PMID: 35806048 PMCID: PMC9266935 DOI: 10.3390/ijms23137029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Although the study on the regulatory mechanism of endothelial differentiation from the perspective of development provides references for endothelial cell (EC) derivation from pluripotent stem cells, incomplete reprogramming and donor-specific epigenetic memory are still thought to be the obstacles of iPSCs for clinical application. Thus, it is necessary to establish a stable iPSC-EC induction system and investigate the regulatory mechanism of endothelial differentiation. Based on a single-layer culture system, we successfully obtained ECs from porcine iPSCs (piPSCs). In vitro, the derived piPSC-ECs formed microvessel-like structures along 3D gelatin scaffolds. Under pathological conditions, the piPSC-ECs functioned on hindlimb ischemia repair by promoting blood vessel formation. To elucidate the molecular events essential for endothelial differentiation in our model, genome-wide transcriptional profile analysis was conducted, and we found that during piPSC-EC derivation, the synthesis and secretion level of TGF-β as well as the phosphorylation level of Smad2/3 changed dynamically. TGF-β-Smad2/3 signaling activation promoted mesoderm formation and prevented endothelial differentiation. Understanding the regulatory mechanism of iPSC-EC derivation not only paves the way for further optimization, but also provides reference for establishing a cardiovascular drug screening platform and revealing the molecular mechanism of endothelial dysfunction.
Collapse
Affiliation(s)
- Yang Yu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
| | - Xuechun Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
| | - Yimei Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
| | - Renyue Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
| | - Hai Li
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China;
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
- Correspondence: (Z.L.); (Y.Z.)
| | - Yu Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.Y.); (X.L.); (Y.L.); (R.W.)
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China;
- Correspondence: (Z.L.); (Y.Z.)
| |
Collapse
|
11
|
Boonkaew B, Suwanpitak S, Pattanapanyasat K, Sermsathanasawadi N, Wattanapanitch M. Efficient generation of endothelial cells from induced pluripotent stem cells derived from a patient with peripheral arterial disease. Cell Tissue Res 2022; 388:89-104. [DOI: 10.1007/s00441-022-03576-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022]
|
12
|
Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions. J Cardiovasc Dev Dis 2021; 8:jcdd8110148. [PMID: 34821701 PMCID: PMC8622843 DOI: 10.3390/jcdd8110148] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great promise for cardiovascular regeneration following ischemic injury. Considerable effort has been made toward the development and optimization of methods to differentiate hiPSCs into vascular cells, such as endothelial and smooth muscle cells (ECs and SMCs). In particular, hiPSC-derived ECs have shown robust potential for promoting neovascularization in animal models of cardiovascular diseases, potentially achieving significant and sustained therapeutic benefits. However, the use of hiPSC-derived SMCs that possess high therapeutic relevance is a relatively new area of investigation, still in the earlier investigational stages. In this review, we first discuss different methodologies to derive vascular cells from hiPSCs with a particular emphasis on the role of key developmental signals. Furthermore, we propose a standardized framework for assessing and defining the EC and SMC identity that might be suitable for inducing tissue repair and regeneration. We then highlight the regenerative effects of hiPSC-derived vascular cells on animal models of myocardial infarction and hindlimb ischemia. Finally, we address several obstacles that need to be overcome to fully implement the use of hiPSC-derived vascular cells for clinical application.
Collapse
|
13
|
Bubb KJ, Harmer JA, Finemore M, Aitken SJ, Ali ZS, Billot L, Chow C, Golledge J, Mister R, Gray MP, Grieve SM, Hamburg N, Keech AC, Patel S, Puttaswamy V, Figtree GA. Protocol for the Stimulating β 3-Adrenergic Receptors for Peripheral Artery Disease (STAR-PAD) trial: a double-blinded, randomised, placebo-controlled study evaluating the effects of mirabegron on functional performance in patients with peripheral arterial disease. BMJ Open 2021; 11:e049858. [PMID: 34588252 PMCID: PMC8479946 DOI: 10.1136/bmjopen-2021-049858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION There is currently only one approved medication effective at improving walking distance in people with intermittent claudication. Preclinical data suggest that the β3-adrenergic receptor agonist (mirabegron) could be repurposed to treat intermittent claudication associated with peripheral artery disease. The aim of the Stimulating β3-Adrenergic Receptors for Peripheral Artery Disease (STAR-PAD) trial is to test whether mirabegron improves walking distance in people with intermittent claudication. METHODS AND ANALYSIS The STAR-PAD trial is a Phase II, multicentre, double-blind, randomised, placebo-controlled trial of mirabegron versus placebo on walking distance in patients with PAD. A total of 120 patients aged ≥40 years with stable PAD and intermittent claudication will be randomly assigned (1:1 ratio) to receive either mirabegron (50 mg orally once a day) or matched placebo, for 12 weeks. The primary endpoint is change in peak walking distance as assessed by a graded treadmill test. Secondary endpoints will include: (i) initial claudication distance; (ii) average daily step count and total step count and (iii) functional status and quality of life assessment. Mechanistic substudies will examine potential effects of mirabegron on vascular function, including brachial artery flow-mediate dilatation; MRI assessment of lower limb blood flow, tissue perfusion and arterial stiffness and numbers and angiogenesis potential of endothelial progenitor cells. Given that mirabegron is safe and clinically available for alternative purposes, a positive study is positioned to immediately impact patient care. ETHICS AND DISSEMINATION The STAR-PAD trial is approved by the Northern Sydney Local Health District Human Research Ethics Committee (HREC/18/HAWKE/50). The study results will be published in peer-reviewed medical or scientific journals and presented at scientific meetings, regardless of the study outcomes. TRIAL REGISTRATION NUMBER ACTRN12619000423112; Results.
Collapse
Affiliation(s)
- Kristen J Bubb
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Jason A Harmer
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Meghan Finemore
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Sarah Joy Aitken
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Zara S Ali
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Laurent Billot
- The George Institute for Global Health, UNSW Sydney, Newtown, New South Wales, Australia
| | - Clara Chow
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- The George Institute for Global Health, UNSW Sydney, Newtown, New South Wales, Australia
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Jonathan Golledge
- Vascular Biology Unit, James Cook University Queensland Research Centre for Peripheral Vascular Disease, Townsville, Queensland, Australia
| | - Rebecca Mister
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Michael P Gray
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Stuart M Grieve
- The Heart Research Institute, Newtown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | - Anthony C Keech
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sanjay Patel
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- The Heart Research Institute, Newtown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Vikram Puttaswamy
- Vascular Surgery, North Shore Private Hospital, Sydney, New South Wales, Australia
| | - Gemma A Figtree
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
14
|
Wingo M, Rafii S. Endothelial reprogramming for vascular regeneration: Past milestones and future directions. Semin Cell Dev Biol 2021; 122:50-55. [PMID: 34548212 DOI: 10.1016/j.semcdb.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Endothelial cells are critical mediators of health and disease. Regenerative medicine techniques that target the endothelium hold vast promise for improving lifespan and quality of life worldwide. Regenerative therapies via induced pluripotent stem cells (IPSCs) have helped demonstrate disease mechanisms, but so far, concerns regarding their function, malignant potential, and expense have limited therapeutic potential. One alternative approach is direct reprogramming of somatic cells, which avoids the pluripotent state and allows for in vivo reprogramming. Transcription factors from endothelial development have yielded essential transcription factors and small molecules that induce endothelial cell fate. Most direct cell reprogramming strategies targeting endothelial cells use ETV2, a pioneer transcription factor to specify endothelial lineage via histone-modifying enzymes. Many different types of starting cells and strategies, including lentiviral transduction, inducing innate immunity, and small molecule signaling have been leveraged for reprogramming. However, so far therapeutic benefit of these strategies remains unproven. Future research will have to solve scalability, safety, and efficacy hurdles before being ready for the clinic. However, researchers have already discovered meaningful insights into disease mechanisms and development through direct reprogramming.
Collapse
Affiliation(s)
- Matthew Wingo
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
15
|
Hywood JD, Sadeghipour S, Clayton ZE, Yuan J, Stubbs C, Wong JWT, Cooke JP, Patel S. Induced endothelial cells from peripheral arterial disease patients and neonatal fibroblasts have comparable angiogenic properties. PLoS One 2021; 16:e0255075. [PMID: 34375370 PMCID: PMC8354451 DOI: 10.1371/journal.pone.0255075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/11/2021] [Indexed: 12/05/2022] Open
Abstract
Induced endothelial cells (iECs) generated from neonatal fibroblasts via transdifferentiation have been shown to have pro-angiogenic properties and are a potential therapy for peripheral arterial disease (PAD). It is unknown if iECs can be generated from fibroblasts collected from PAD patients and whether these cells are pro-angiogenic. In this study fibroblasts were collected from four PAD patients undergoing carotid endarterectomies. These cells, and neonatal fibroblasts, were transdifferentiated into iECs using modified mRNA. Endothelial phenotype and pro-angiogenic cytokine secretion were investigated. NOD-SCID mice underwent surgery to induce hindlimb ischaemia in a murine model of PAD. Mice received intramuscular injections with either control vehicle, or 1 × 106 neonatal-derived or 1 × 106 patient-derived iECs. Recovery in perfusion to the affected limb was measured using laser Doppler scanning. Perfusion recovery was enhanced in mice treated with neonatal-derived iECs and in two of the three patient-derived iEC lines investigated in vivo. Patient-derived iECs can be successfully generated from PAD patients and for specific patients display comparable pro-angiogenic properties to neonatal-derived iECs.
Collapse
Affiliation(s)
- Jack D. Hywood
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | | | - Zoe E. Clayton
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Jun Yuan
- Heart Research Institute, Newtown, NSW, Australia
| | - Colleen Stubbs
- RNACore, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Jack W. T. Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Sanjay Patel
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
16
|
Roux BM, Vaicik MK, Shrestha B, Montelongo S, Stojkova K, Yang F, Guda T, Cinar A, Brey EM. Induced Pluripotent Stem Cell-Derived Endothelial Networks Accelerate Vascularization But Not Bone Regeneration. Tissue Eng Part A 2021; 27:940-961. [PMID: 32924856 PMCID: PMC8336421 DOI: 10.1089/ten.tea.2020.0200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022] Open
Abstract
Vascularization is critical for engineering mineralized tissues. It has been previously shown that biomaterials containing preformed endothelial networks anastomose to host vasculature following implantation. However, the networks alone may not increase regeneration. In addition, a clinically applicable source of cells for vascularization is needed. In this study, vascular networks were generated from endothelial cells (ECs) derived from human induced pluripotent stem cells (iPSCs). Network formation by iPSC-ECs within fibrin gels was investigated in a mesenchymal stem cells (MSCs) coculture spheroid model. Statistical design of experiments technique was evaluated for its predicting capability during the optimization of experimental parameters. The prevascularized units were combined with hydroxyapatite nanoparticles to develop a vascularized composite hydrogel that was implanted in a rodent critical-sized cranial defect model. Immunohistological staining for human-specific CD31 at week 1 indicated the presence and maintenance of the implanted vessels. At 8 weeks, the prevascularized systems resulted in higher vessel density over MSC-only scaffolds. The implanted vessels appeared to establish flow with host vasculature. While there was a slight increase in bone volume in the prevascularized bone construct compared to MSC-only bone constructs, there was not a profound increase in bone regeneration. These results show that scaffolds with network structures can be generated from ECs derived from iPSC and that the networks survive and inosculate with the host postimplantation in a bone model. Impact statement Vascularization is critical for engineering bone. Prevascularized scaffolds have been shown to improve postimplantation vascularization. Herein, vascularized networks were generated from induced pluripotent cells derived from endothelial cells. These vascularized units were combined with a fibrin/hydroxyapatite scaffold to develop a prevascularized construct for bone regeneration. Implantation of these scaffolds in a small animal cranial defect model resulted in network inosculation and increased vascularization, but exhibited only a limited effect on bone formation. This study provides insight into the challenges of generating vascularized bone.
Collapse
Affiliation(s)
- Brianna M. Roux
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
- Department of Research Service, Edward Hines, Jr. VA Hospital, Hines, Illinois, USA
| | - Marcella K. Vaicik
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
- Department of Research Service, Edward Hines, Jr. VA Hospital, Hines, Illinois, USA
| | - Binita Shrestha
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Sergio Montelongo
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Katerina Stojkova
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Feipeng Yang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Ali Cinar
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Eric M. Brey
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
- Department of Research Service, Edward Hines, Jr. VA Hospital, Hines, Illinois, USA
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
17
|
Alsaigh T, Di Bartolo BA, Mulangala J, Figtree GA, Leeper NJ. Bench-to-Bedside in Vascular Medicine: Optimizing the Translational Pipeline for Patients With Peripheral Artery Disease. Circ Res 2021; 128:1927-1943. [PMID: 34110900 PMCID: PMC8208504 DOI: 10.1161/circresaha.121.318265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peripheral arterial disease is a growing worldwide problem with a wide spectrum of clinical severity and is projected to consume >$21 billion per year in the United States alone. While vascular researchers have brought several therapies to the clinic in recent years, few of these approaches have leveraged advances in high-throughput discovery screens, novel translational models, or innovative trial designs. In the following review, we discuss recent advances in unbiased genomics and broader omics technology platforms, along with preclinical vascular models designed to enhance our understanding of disease pathobiology and prioritize targets for additional investigation. Furthermore, we summarize novel approaches to clinical studies in subjects with claudication and ischemic ulceration, with an emphasis on streamlining and accelerating bench-to-bedside translation. By providing a framework designed to enhance each aspect of future clinical development programs, we hope to enrich the pipeline of therapies that may prevent loss of life and limb for those with peripheral arterial disease.
Collapse
Affiliation(s)
- Tom Alsaigh
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Belinda A. Di Bartolo
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | | | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
18
|
Abstract
The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation. Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms, and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease. Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic angiogenesis and arteriogenesis for PAD.
Collapse
Affiliation(s)
- Brian H Annex
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University (B.H.A.)
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.)
| |
Collapse
|
19
|
Bubb KJ, Ravindran D, Cartland SP, Finemore M, Clayton ZE, Tsang M, Tang O, Kavurma MM, Patel S, Figtree GA. β 3 Adrenergic Receptor Stimulation Promotes Reperfusion in Ischemic Limbs in a Murine Diabetic Model. Front Pharmacol 2021; 12:666334. [PMID: 33967810 PMCID: PMC8100512 DOI: 10.3389/fphar.2021.666334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Aims/Hypothesis: Peripheral arterial disease (PAD) is a major burden, resulting in limb claudication, repeated surgical interventions and amputation. There is an unmet need for improved medical management of PAD that improves quality of life, maintains activities of daily life and reduces complications. Nitric oxide (NO)/redox balance is a key regulator of angiogenesis. We have previously shown beneficial effects of a β3 adrenergic receptor (β3AR) agonist on NO/redox balance. We hypothesized that β3AR stimulation would have therapeutic potential in PAD by promoting limb angiogenesis. Methods: The effect of the β3AR agonist CL 316,243 (1–1,000 nmol/L in vitro, 1 mg/kg/day s. c) was tested in established angiogenesis assays with human endothelial cells and patient-derived endothelial colony forming cells. Post-ischemia reperfusion was determined in streptozotocin and/or high fat diet-induced diabetic and non-diabetic mice in vivo using the hind limb ischemia model. Results: CL 316,243 caused accelerated recovery from hind limb ischemia in non-diabetic and type 1 and 2 diabetic mice. Increased eNOS activity and decreased superoxide generation were detected in hind limb ischemia calf muscle from CL 316, 243 treated mice vs. controls. The protective effect of CL 316,243 in diabetic mice was associated with >50% decreases in eNOS glutathionylation and nitrotyrosine levels. The β3AR agonist directly promoted angiogenesis in endothelial cells in vitro. These pro-angiogenic effects were β3AR and NOS-dependent. Conclusion/Interpretation:β3AR stimulation increased angiogenesis in diabetic ischemic limbs, with demonstrable improvements in NO/redox balance and angiogenesis elicited by a selective agonist. The orally available β3AR agonist, Mirabegron, used for overactive bladder syndrome, makes translation to a clinical trial by repurposing of a β3AR agonist to target PAD immediately feasible.
Collapse
Affiliation(s)
- Kristen J Bubb
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia.,Department of Physiology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Dhanya Ravindran
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Siân P Cartland
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Meghan Finemore
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Zoe E Clayton
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Michael Tsang
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Owen Tang
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Mary M Kavurma
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Sanjay Patel
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Gemma A Figtree
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
20
|
Gao X, Gao M, Gorecka J, Langford J, Liu J, Luo J, Taniguchi R, Matsubara Y, Liu H, Guo L, Gu Y, Qyang Y, Dardik A. Human-Induced Pluripotent Stem-Cell-Derived Smooth Muscle Cells Increase Angiogenesis to Treat Hindlimb Ischemia. Cells 2021; 10:792. [PMID: 33918299 PMCID: PMC8066461 DOI: 10.3390/cells10040792] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSC) represent an innovative, somatic cell-derived, easily obtained and renewable stem cell source without considerable ethical issues. iPSC and their derived cells may have enhanced therapeutic and translational potential compared with other stem cells. We previously showed that human iPSC-derived smooth muscle cells (hiPSC-SMC) promote angiogenesis and wound healing. Accordingly, we hypothesized that hiPSC-SMC may be a novel treatment for human patients with chronic limb-threatening ischemia who have no standard options for therapy. We determined the angiogenic potential of hiPSC-SMC in a murine hindlimb ischemia model. hiPSC-SMC were injected intramuscularly into nude mice after creation of hindlimb ischemia. Functional outcomes and perfusion were measured using standardized scores, laser Doppler imaging, microCT, histology and immunofluorescence. Functional outcomes and blood flow were improved in hiPSC-SMC-treated mice compared with controls (Tarlov score, p < 0.05; Faber score, p < 0.05; flow, p = 0.054). hiPSC-SMC-treated mice showed fewer gastrocnemius fibers (p < 0.0001), increased fiber area (p < 0.0001), and enhanced capillary density (p < 0.01); microCT showed more arterioles (<96 μm). hiPSC-SMC treatment was associated with fewer numbers of macrophages, decreased numbers of M1-type (p < 0.05) and increased numbers of M2-type macrophages (p < 0.0001). Vascular endothelial growth factor (VEGF) expression in ischemic limbs was significantly elevated with hiPSC-SMC treatment (p < 0.05), and inhibition of VEGFR-2 with SU5416 was associated with fewer capillaries in hiPSC-SMC-treated limbs (p < 0.0001). hiPSC-SMC promote VEGF-mediated angiogenesis, leading to improved hindlimb ischemia. Stem cell therapy using iPSC-derived cells may represent a novel and potentially translatable therapy for limb-threatening ischemia.
Collapse
Affiliation(s)
- Xixiang Gao
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University and Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China; (X.G.); (L.G.); (Y.G.)
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - Mingjie Gao
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Vascular Ultrasound, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jolanta Gorecka
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - John Langford
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - Jia Liu
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - Jiesi Luo
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Ryosuke Taniguchi
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - Yutaka Matsubara
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Surgery and Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hao Liu
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
| | - Lianrui Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University and Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China; (X.G.); (L.G.); (Y.G.)
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University and Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China; (X.G.); (L.G.); (Y.G.)
| | - Yibing Qyang
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Alan Dardik
- Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519, USA; (M.G.); (J.G.); (J.L.); (J.L.); (J.L.); (R.T.); (Y.M.); (H.L.); (Y.Q.)
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Surgery, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
21
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
22
|
Li C, Kitzerow O, Nie F, Dai J, Liu X, Carlson MA, Casale GP, Pipinos II, Li X. Bioengineering strategies for the treatment of peripheral arterial disease. Bioact Mater 2021; 6:684-696. [PMID: 33005831 PMCID: PMC7511653 DOI: 10.1016/j.bioactmat.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by narrowing and occlusion of arteries supplying the lower extremities. Approximately 200 million people worldwide are affected by PAD. The current standard of operative care is open or endovascular revascularization in which blood flow restoration is the goal. However, many patients are not appropriate candidates for these treatments and are subject to continuous ischemia of their lower limbs. Current research in the therapy of PAD involves developing modalities that induce angiogenesis, but the results of simple cell transplantation or growth factor delivery have been found to be relatively poor mainly due to difficulties in stem cell retention and survival and rapid diffusion and enzymolysis of growth factors following injection of these agents in the affected tissues. Biomaterials, including hydrogels, have the capability to protect stem cells during injection and to support cell survival. Hydrogels can also provide a sustained release of growth factors at the injection site. This review will focus on biomaterial systems currently being investigated as carriers for cell and growth factor delivery, and will also discuss biomaterials as a potential stand-alone method for the treatment of PAD. Finally, the challenges of development and use of biomaterials systems for PAD treatment will be reviewed.
Collapse
Affiliation(s)
- Cui Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jingxuan Dai
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Omaha VA Medical Center, Omaha, NE, 68105, United States
| | - George P. Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| |
Collapse
|
23
|
Nour S, Imani R, Chaudhry GR, Sharifi AM. Skin wound healing assisted by angiogenic targeted tissue engineering: A comprehensive review of bioengineered approaches. J Biomed Mater Res A 2020; 109:453-478. [PMID: 32985051 DOI: 10.1002/jbm.a.37105] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/23/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022]
Abstract
Skin injuries and in particular, chronic wounds, are one of the major prevalent medical problems, worldwide. Due to the pivotal role of angiogenesis in tissue regeneration, impaired angiogenesis can cause several complications during the wound healing process and skin regeneration. Therefore, induction or promotion of angiogenesis can be considered as a promising approach to accelerate wound healing. This article presents a comprehensive overview of current and emerging angiogenesis induction methods applied in several studies for skin regeneration, which are classified into the cell, growth factor, scaffold, and biological/chemical compound-based strategies. In addition, the advantages and disadvantages of these angiogenic strategies along with related research examples are discussed in order to demonstrate their potential in the treatment of wounds.
Collapse
Affiliation(s)
- Shirin Nour
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - G Rasul Chaudhry
- OU-WB Institute for Stem Cell and Regenerative Medicine, Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Group (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Gorecka J, Gao X, Fereydooni A, Dash BC, Luo J, Lee SR, Taniguchi R, Hsia HC, Qyang Y, Dardik A. Induced pluripotent stem cell-derived smooth muscle cells increase angiogenesis and accelerate diabetic wound healing. Regen Med 2020; 15:1277-1293. [PMID: 32228292 PMCID: PMC7304438 DOI: 10.2217/rme-2019-0086] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To assess the potential of human induced pluripotent stem cell-derived smooth muscle cells (hiPSC-SMC) to accelerate diabetic wound healing. Methods: hiPSC-SMC were embedded in 3D collagen scaffolds and cultured in vitro for 72 h; scaffolds were then applied to diabetic, nude mouse, splinted back wounds to assess in vivo healing. Cultured medium after scaffold incubation was collected and analyzed for expression of pro-angiogenic cytokines. Results: hiPSC-SMC secrete increased concentration of pro-angiogenic cytokines, compared with murine adipose derived stem cells. Delivery of hiPSC-SMC-containing collagen scaffolds accelerates diabetic wound healing and is associated with an increased number of total and M2 type macrophages. Conclusion: hiPSC-SMC promote angiogenesis and accelerate diabetic wound healing, making them a promising new candidate for treatment of diabetic wounds.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Xixiang Gao
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA.,Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Arash Fereydooni
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Biraja C Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA.,Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519 USA.,Department of Pathology, YaleUniversity, New Haven, CT 06520, USA
| | - Shin Rong Lee
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Ryosuke Taniguchi
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA.,Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519 USA.,Department of Pathology, YaleUniversity, New Haven, CT 06520, USA
| | - Alan Dardik
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA.,Section of Vascular & Endovascular Surgery, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
25
|
Cell-modified bioprinted microspheres for vascular regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110896. [PMID: 32409053 DOI: 10.1016/j.msec.2020.110896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 03/21/2020] [Accepted: 03/21/2020] [Indexed: 01/28/2023]
Abstract
Cell therapy is a promising strategy in which living cells or cellular materials are delivered to treat a variety of diseases. Here, we developed an electrospray bioprinting method to rapidly generate cell-laden hydrogel microspheres, which limit the migration of the captured cells and provide an immunologically privileged microenvironment for cell survival in vivo. Currently, therapeutic angiogenesis aims to induce collateral vessel formation after limb ischemia. However, the clinical application of gene and cell therapy has been impeded by concerns regarding its inefficacy, as well as the associated risk of immunogenicity and oncogenicity. In this study, hydrogel microspheres encapsulating VEGF-overexpressing HEK293T cells showed good safety via subcutaneously injecting into male C57BL/6 mice. In addition, these cell-modified microspheres effectively promoted angiogenesis in a mouse hind-limb ischemia model. Therefore, we demonstrated the great therapeutic potential of this approach to induce angiogenesis in limb ischemia, indicating that bioprinting has a bright future in cell therapy.
Collapse
|
26
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
27
|
Palladino A, Mavaro I, Pizzoleo C, De Felice E, Lucini C, de Girolamo P, Netti PA, Attanasio C. Induced Pluripotent Stem Cells as Vasculature Forming Entities. J Clin Med 2019; 8:E1782. [PMID: 31731464 PMCID: PMC6912734 DOI: 10.3390/jcm8111782] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 01/06/2023] Open
Abstract
Tissue engineering (TE) pursues the ambitious goal to heal damaged tissues. One of the most successful TE approaches relies on the use of scaffolds specifically designed and fabricated to promote tissue growth. During regeneration the guidance of biological events may be essential to sustain vasculature neoformation inside the engineered scaffold. In this context, one of the most effective strategies includes the incorporation of vasculature forming cells, namely endothelial cells (EC), into engineered constructs. However, the most common EC sources currently available, intended as primary cells, are affected by several limitations that make them inappropriate to personalized medicine. Human induced Pluripotent Stem Cells (hiPSC), since the time of their discovery, represent an unprecedented opportunity for regenerative medicine applications. Unfortunately, human induced Pluripotent Stem Cells-Endothelial Cells (hiPSC-ECs) still display significant safety issues. In this work, we reviewed the most effective protocols to induce pluripotency, to generate cells displaying the endothelial phenotype and to perform an efficient and safe cell selection. We also provide noteworthy examples of both in vitro and in vivo applications of hiPSC-ECs in order to highlight their ability to form functional blood vessels. In conclusion, we propose hiPSC-ECs as the preferred source of endothelial cells currently available in the field of personalized regenerative medicine.
Collapse
Affiliation(s)
- Antonio Palladino
- CESMA—Centro Servizi Metrologici e Tecnologici Avanzati, University of Naples Federico II, 80146 Naples, Italy
| | - Isabella Mavaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
| | - Carmela Pizzoleo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
| | - Elena De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
| | - Paolo A. Netti
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Chiara Attanasio
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
28
|
Browne S, Healy KE. Matrix-assisted cell transplantation for tissue vascularization. Adv Drug Deliv Rev 2019; 146:155-169. [PMID: 30605738 DOI: 10.1016/j.addr.2018.12.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/30/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022]
Abstract
Cell therapy offers much promise for the treatment of ischemic diseases by augmenting tissue vasculogenesis. Matrix-assisted cell transplantation (MACT) has been proposed as a solution to enhance cell survival and integration with host tissue following transplantation. By designing semi synthetic matrices (sECM) with the correct physical and biochemical signals, encapsulated cells are directed towards a more angiogenic phenotype. In this review, we describe the choice of cells suitable for pro-angiogenic therapies, the properties that should be considered when designing sECM for transplantation and their relative importance. Pre-clinical models where MACT has been successfully applied to promote angiogenesis are reviewed to show the great potential of this strategy to treat ischemic conditions.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA; Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
29
|
Gorecka J, Kostiuk V, Fereydooni A, Gonzalez L, Luo J, Dash B, Isaji T, Ono S, Liu S, Lee SR, Xu J, Liu J, Taniguchi R, Yastula B, Hsia HC, Qyang Y, Dardik A. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res Ther 2019; 10:87. [PMID: 30867069 PMCID: PMC6416973 DOI: 10.1186/s13287-019-1185-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Wound healing is the physiologic response to a disruption in normal skin architecture and requires both spatial and temporal coordination of multiple cell types and cytokines. This complex process is prone to dysregulation secondary to local and systemic factors such as ischemia and diabetes that frequently lead to chronic wounds. Chronic wounds such as diabetic foot ulcers are epidemic with great cost to the healthcare system as they heal poorly and recur frequently, creating an urgent need for new and advanced therapies. Stem cell therapy is emerging as a potential treatment for chronic wounds, and adult-derived stem cells are currently employed in several commercially available products; however, stem cell therapy is limited by the need for invasive harvesting techniques, immunogenicity, and limited cell survival in vivo. Induced pluripotent stem cells (iPSC) are an exciting cell type with enhanced therapeutic and translational potential. iPSC are derived from adult cells by in vitro induction of pluripotency, obviating the ethical dilemmas surrounding the use of embryonic stem cells; they are harvested non-invasively and can be transplanted autologously, reducing immune rejection; and iPSC are the only cell type capable of being differentiated into all of the cell types in healthy skin. This review focuses on the use of iPSC in animal models of wound healing including limb ischemia, as well as their limitations and methods aimed at improving iPSC safety profile in an effort to hasten translation to human studies.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Valentyna Kostiuk
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Biraja Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shin Rong Lee
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jianbiao Xu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jia Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Bogdan Yastula
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, USA.,Department of Pathology, Yale University, New Haven, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA.
| |
Collapse
|
30
|
Dong X, Li H, E L, Cao J, Guo B. Bioceramic akermanite enhanced vascularization and osteogenic differentiation of human induced pluripotent stem cells in 3D scaffolds in vitro and vivo. RSC Adv 2019; 9:25462-25470. [PMID: 35530104 PMCID: PMC9070079 DOI: 10.1039/c9ra02026h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/19/2019] [Indexed: 01/10/2023] Open
Abstract
A growing number of studies suggest that the modulation of cell differentiation by biomaterials is critical for tissue engineering. In previous work, we demonstrated that human induced pluripotent stem cells (iPSCs) are remarkably promising seed cells for bone tissue engineering. In addition, we found that the ionic products of akermanite (Aker) are potential inducers of osteogenic differentiation of iPSCs. Furthermore, composite scaffolds containing polymer and bioceramics have more interesting properties compared to pure bioceramic scaffolds for bone tissue engineering. The characteristic of model biomaterials in bone tissue engineering is their ability to control the osteogenic differentiation of stem cells and simultaneously induce the angiogenesis of endothelia cells. Thus, this study aimed at investigating the effects of poly(lactic-co-glycolic acid)/Aker (PLGA-Aker) composite scaffolds on angiogenic and osteogenic differentiation of human iPSCs in order to optimize the scaffold compositions. The results from Alizarin Red S staining, qRT-PCR analysis of osteogenic genes (BMP2, RUNX2, ALP, COL1 and OCN) and angiogenic genes (VEGF and CD31) demonstrated that PLGA/Aker composite scaffolds containing 10% Aker exhibited the highest stimulatory effects on the osteogenic and angiogenic differentiation of human iPSCs among all scaffolds. After the scaffolds were implanted in nu/nu mice subcutaneous pockets and calvarial defects, H&E staining, BSP immunostaining, qRT-PCR analysis and micro-CT analysis (BMD, BV/TV) indicated that PLGA + 10% Aker scaffolds enhanced the vascularization and osteogenic differentiation of human iPSCs and stimulated the repair of bone defects. Taken together, our work indicated that combining scaffolds containing silicate bioceramic Aker and human iPSCs is a promising approach for the enhancement of bone regeneration. Bioceramics akermanite enhanced vascularization and osteogenic differentiation of human iPSCs in 3D scaffolds in vitro and vivo.![]()
Collapse
Affiliation(s)
- Xixi Dong
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Haiyan Li
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Lingling E
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Junkai Cao
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| | - Bin Guo
- Stomatology Department
- General Hospital of Chinese PLA
- Beijing 100853
- China
| |
Collapse
|
31
|
Induced pluripotent stem cell-derived endothelial cells promote angiogenesis and accelerate wound closure in a murine excisional wound healing model. Biosci Rep 2018; 38:BSR20180563. [PMID: 29976773 PMCID: PMC6066657 DOI: 10.1042/bsr20180563] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/04/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022] Open
Abstract
Chronic wounds are a major complication in patients with cardiovascular diseases. Cell therapies have shown potential to stimulate wound healing, but clinical trials using adult stem cells have been tempered by limited numbers of cells and invasive procurement procedures. Induced pluripotent stem cells (iPSCs) have several advantages of other cell types, for example they can be generated in abundance from patients’ somatic cells (autologous) or those from a matched donor. iPSCs can be efficiently differentiated to functional endothelial cells (iPSC-ECs). Here, we used a murine excisional wound model to test the pro-angiogenic properties of iPSC-ECs in wound healing. Two full-thickness wounds were made on the dorsum of NOD-SCID mice and splinted. iPSC-ECs (5 × 105) were topically applied to one wound, with the other serving as a control. Treatment with iPSC-ECs significantly increased wound perfusion and accelerated wound closure. Expression of endothelial cell (EC) surface marker, platelet endothelial cell adhesion molecule (PECAM-1) (CD31), and pro-angiogenic EC receptor, Tie1, mRNA was up-regulated in iPSC-EC treated wounds at 7 days post-wounding. Histological analysis of wound sections showed increased capillary density in iPSC-EC wounds at days 7 and 14 post-wounding, and increased collagen content at day 14. Anti-GFP fluorescence confirmed presence of iPSC-ECs in the wounds. Bioluminescent imaging (BLI) showed progressive decline of iPSC-ECs over time, suggesting that iPSC-ECs are acting primarily through short-term paracrine effects. These results highlight the pro-regenerative effects of iPSC-ECs and demonstrate that they are a promising potential therapy for intractable wounds.
Collapse
|
32
|
Tan RP, Chan AHP, Lennartsson K, Miravet MM, Lee BSL, Rnjak-Kovacina J, Clayton ZE, Cooke JP, Ng MKC, Patel S, Wise SG. Integration of induced pluripotent stem cell-derived endothelial cells with polycaprolactone/gelatin-based electrospun scaffolds for enhanced therapeutic angiogenesis. Stem Cell Res Ther 2018; 9:70. [PMID: 29562916 PMCID: PMC5863387 DOI: 10.1186/s13287-018-0824-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/19/2018] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Induced pluripotent stem-cell derived endothelial cells (iPSC-ECs) can be generated from any somatic cell and their iPSC sources possess unlimited self-renewal. Previous demonstration of their proangiogenic activity makes them a promising cell type for treatment of ischemic injury. As with many other stem cell approaches, the low rate of in-vivo survival has been a major limitation to the efficacy of iPSC-ECs to date. In this study, we aimed to increase the in-vivo lifetime of iPSC-ECs by culturing them on electrospun polycaprolactone (PCL)/gelatin scaffolds, before quantifying the subsequent impact on their proangiogenic function. METHODS iPSC-ECs were isolated and stably transfected with a luciferase reporter to facilitate quantification of cell numbers and non-invasive imaging in-vivo PCL/gelatin scaffolds were engineered using electrospinning to obtain woven meshes of nanofibers. iPSC-ECs were cultured on scaffolds for 7 days. Subsequently, cell growth and function were assessed in vitro followed by implantation in a mouseback subcutaneous model for 7 days. RESULTS Using a matrix of conditions, we found that scaffold blends with ratios of PCL:gelatin of 70:30 (PG73) spun at high flow rates supported the greatest levels of iPSC-EC growth, retention of phenotype, and function in vitro. Implanting iPSC-ECs seeded on PG73 scaffolds in vivo improved their survival up to 3 days, compared to cells directly injected into control wounds, which were no longer observable within 1 h. Enhanced engraftment improved blood perfusion, observed through non-invasive laser Doppler imaging. Immunohistochemistry revealed a corresponding increase in host angiogenic mechanisms characterized by the enhanced recruitment of macrophages and the elevated expression of proangiogenic cytokines vascular endothelial growth factor and placental growth factor. CONCLUSIONS Knowledge of these mechanisms combined with a deeper understanding of the scaffold parameters influencing this function provides the groundwork for optimizing future iPSC-EC therapies utilizing engraftment platforms. The development of combined scaffold and iPSC-EC therapies could ultimately improve therapeutic angiogenesis and the treatment of ischemic injury.
Collapse
Affiliation(s)
- Richard P Tan
- The Heart Research Institute, Sydney, NSW, 2042, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Alex H P Chan
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | | | | | - Bob S L Lee
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zoe E Clayton
- The Heart Research Institute, Sydney, NSW, 2042, Australia
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Martin K C Ng
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, 2042, Australia
| | - Sanjay Patel
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, 2042, Australia
| | - Steven G Wise
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
33
|
Foster AA, Dewi RE, Cai L, Hou L, Strassberg Z, Alcazar C, Heilshorn SC, Huang NF. Protein-engineered hydrogels enhance the survival of induced pluripotent stem cell-derived endothelial cells for treatment of peripheral arterial disease. Biomater Sci 2018; 6:614-622. [PMID: 29406542 PMCID: PMC5829050 DOI: 10.1039/c7bm00883j] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A key feature of peripheral arterial disease (PAD) is damage to endothelial cells (ECs), resulting in lower limb pain and restricted blood flow. Recent preclinical studies demonstrate that the transplantation of ECs via direct injection into the affected limb can result in significantly improved blood circulation. Unfortunately, the clinical application of this therapy has been limited by low cell viability and poor cell function. To address these limitations we have developed an injectable, recombinant hydrogel, termed SHIELD (Shear-thinning Hydrogel for Injectable Encapsulation and Long-term Delivery) for cell transplantation. SHIELD provides mechanical protection from cell membrane damage during syringe flow. Additionally, secondary in situ crosslinking provides a reinforcing network to improve cell retention, thereby augmenting the therapeutic benefit of cell therapy. In this study, we demonstrate the improved acute viability of human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) following syringe injection delivery in SHIELD, compared to saline. Using a murine hind limb ischemia model of PAD, we demonstrate enhanced iPSC-EC retention in vivo and improved neovascularization of the ischemic limb based on arteriogenesis following transplantation of iPSC-ECs delivered in SHIELD.
Collapse
Affiliation(s)
- Abbygail A. Foster
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Lei Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Luqia Hou
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Cynthia Alcazar
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ngan F. Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Cardiothoracic Surgery, Stanford, CA, USA
| |
Collapse
|
34
|
A Molecular and Clinical Review of Stem Cell Therapy in Critical Limb Ischemia. Stem Cells Int 2017; 2017:3750829. [PMID: 29358955 PMCID: PMC5735649 DOI: 10.1155/2017/3750829] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022] Open
Abstract
Peripheral artery disease (PAD) is one of the major vascular complications in individuals suffering from diabetes and in the elderly that can progress to critical limb ischemia (CLI), portending significant burden in terms of patient morbidity and mortality. Over the last two decades, stem cell therapy (SCT) has risen as an attractive alternative to traditional surgical and/or endovascular revascularization to treat this disorder. The primary benefit of SCT is to induce therapeutic neovascularization and promote collateral vessel formation to increase blood flow in the ischemic limb and soft tissue. Existing evidence provides a solid rationale for ongoing in-depth studies aimed at advancing current SCT that may change the way PAD/CLI patients are treated.
Collapse
|