1
|
Fan C, He J, Xu S, Yan J, Jin L, Dai J, Hu B. Advances in biomaterial-based cardiac organoids. BIOMATERIALS ADVANCES 2023; 153:213502. [PMID: 37352743 DOI: 10.1016/j.bioadv.2023.213502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular disease (CVD) is one of the important causes of death worldwide. The incidence and mortality rates are increasing annually with the intensification of social aging. The efficacy of drug therapy is limited in individuals suffering from severe heart failure due to the inability of myocardial cells to undergo regeneration and the challenging nature of cardiac tissue repair following injury. Consequently, surgical transplantation stands as the most efficient approach for treatment. Nevertheless, the shortage of donors and the considerable number of heart failure patients worldwide, estimated at 26 million, results in an alarming treatment deficit, with only around 5000 heart transplants feasible annually. The existing major alternatives, such as mechanical or xenogeneic hearts, have significant flaws, such as high cost and rejection, and are challenging to implement for large-scale, long-term use. An organoid is a three-dimensional (3D) cell tissue that mimics the characteristics of an organ. The critical application has been rated in annual biotechnology by authoritative journals, such as Science and Cell. Related industries have achieved rapid growth in recent years. Based on this technology, cardiac organoids are expected to pave the way for viable heart repair and treatment and play an essential role in pathological research, drug screening, and other areas. This review centers on the examination of biomaterials employed in cardiac repair, strategies employed for the reconstruction of cardiac structure and function, clinical investigations pertaining to cardiac repair, and the prospective applications of cardiac organoids. From basic research to clinical practice, the current status, latest progress, challenges, and prospects of biomaterial-based cardiac repair are summarized and discussed, providing a reference for future exploration and development of cardiac regeneration strategies.
Collapse
Affiliation(s)
- Caixia Fan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jiaxiong He
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Sijia Xu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China
| | - Junyan Yan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Lifang Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China.
| | - Baowei Hu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
2
|
Zhang S, Ding Y, Feng F, Gao Y. The role of blood CXCL12 level in prognosis of coronary artery disease: A meta-analysis. Front Cardiovasc Med 2022; 9:938540. [PMID: 35966557 PMCID: PMC9363627 DOI: 10.3389/fcvm.2022.938540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
Objective The role of C-X-C motif chemokine 12 (CXCL12) in atherosclerotic cardiovascular diseases (ASCVDs) has emerged as one of the research hotspots in recent years. Studies reported that the higher blood CXCL12 level was associated with increased major adverse cardiovascular events (MACEs), but the results were inconsistent. The objective of this study was to clarify the prognostic value of the blood CXCL12 level in patients with coronary artery disease (CAD) through meta-analysis. Methods All related studies about the association between the blood CXCL12 level and the prognosis of CAD were comprehensively searched and screened according to inclusion criteria and exclusion criteria. The quality of the included literature was evaluated using the Newcastle-Ottawa Scale (NOS). The heterogeneity test was conducted, and the pooled hazard risk (HR) or the odds ratio (OR) with a 95% confidence interval (CI) was calculated using the fixed-effect or random-effects model accordingly. Publication bias was evaluated using Begg's funnel plot and Egger's test. Sensitivity analysis and subgroup analysis were also conducted. Results A total of 12 original studies with 2,959 CAD subjects were included in the final data combination. The pooled data indicated a significant association between higher CXCL12 levels and MACEs both in univariate analysis (HR 5.23, 95% CI 2.48–11.04) and multivariate analysis (HR 2.53, 95% CI 2.03–3.16) in the CXCL12 level as the category variable group. In the CXCL12 level as the continuous variable group, the result also indicated that the higher CXCL12 level significantly predicted future MACEs (multivariate OR 1.55, 95% CI 1.02–2.35). Subgroup analysis of the CXCL12 level as the category variable group found significant associations in all acute coronary syndrome (ACS) (univariate HR 9.72, 95% CI 4.69–20.15; multivariate HR 2.47, 95% CI 1.79–3.40), non-ACS (univariate HR 2.73, 95% CI 1.65–4.54; multivariate HR 3.49, 95% CI 1.66–7.33), Asian (univariate HR 7.43, 95% CI 1.70–32.49; multivariate HR 2.21, 95% CI 1.71–2.85), Caucasian (univariate HR 3.90, 95% CI 2.73–5.57; multivariate HR 3.87, 95% CI 2.48–6.04), short-term (univariate HR 9.36, 95% CI 4.10–21.37; multivariate HR 2.72, 95% CI 1.97–3.76), and long-term (univariate HR 2.86, 95% CI 1.62–5.04; multivariate HR 2.38, 95% CI 1.76–3.22) subgroups. Subgroup analysis of the CXCL12 level as the continuous variable group found significant associations in non-ACS (multivariate OR 1.53, 95% CI 1.23–1.92), Caucasian (multivariate OR 3.83, 95% CI 1.44–10.19), and long-term (multivariate OR 1.62, 95% CI 1.37–1.93) subgroups, but not in ACS (multivariate OR 1.36, 95% CI 0.67–2.75), Asian (multivariate OR 1.40, 95% CI 0.91–2.14), and short-term (multivariate OR 1.16, 95% CI 0.28–4.76) subgroups. No significant publication bias was found in this meta-analysis. Conclusion The higher blood CXCL12 level is associated with increased MACEs in patients with CAD, and the blood CXCL12 level may serve as an important prognostic index for CAD. Integrating the blood CXCL12 level into CAD risk assessment tools may provide more comprehensive messages for evaluating and managing patients with CAD.
Collapse
Affiliation(s)
- Shunrong Zhang
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Ding
- Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Feng
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Gao
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Yue Gao
| |
Collapse
|
3
|
Elde S, Wang H, Woo YJ. Navigating the Crossroads of Cell Therapy and Natural Heart Regeneration. Front Cell Dev Biol 2021; 9:674180. [PMID: 34046410 PMCID: PMC8148343 DOI: 10.3389/fcell.2021.674180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite significant advances in our understanding of the disease and its treatment. Consequently, the therapeutic potential of cell therapy and induction of natural myocardial regeneration have stimulated a recent surge of research and clinical trials aimed at addressing this challenge. Recent developments in the field have shed new light on the intricate relationship between inflammation and natural regeneration, an intersection that warrants further investigation.
Collapse
Affiliation(s)
- Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
4
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
5
|
Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. Stem cell therapy in the heart: Biomaterials as a key route. Tissue Cell 2021; 71:101504. [PMID: 33607524 DOI: 10.1016/j.tice.2021.101504] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are one of the main concerns, nowadays causing a high rate of mortality in the world. The majority of conventional treatment protects the heart from failure progression. As a novel therapeutic way, Regenerative medicine in the heart includes cellular and noncellular approaches. Despite the irrefutable privileges of noncellular aspects such as administration of exosomes, utilizing of miRNAs, and growth factors, they cannot reverse necrotic or ischemic myocardium, hence recruiting of stem cells to help regenerative therapy in the heart seems indispensable. Stem cell lineages are varied and divided into two main groups namely pluripotent and adult stem cells. Not only has each of which own regenerative capacity, benefits, and drawbacks, but their turnover also close correlates with the target organ and/or tissue as well as the stage and level of failure. In addition to inefficient tissue integration due to the defects in delivering methods and poor retention of transplanted cells, the complexity of the heart and its movement also make more rigorous the repair process. Hence, utilizing biomaterials can make a key route to tackle such obstacles. In this review, we evaluate some natural products which can help stem cells in regenerative medicine of the cardiovascular system.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad, Iran; Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Wu Q, Qi B, Duan X, Ming X, Yan F, He Y, Bu X, Sun S, Zhu H. MicroRNA-126 enhances the biological function of endothelial progenitor cells under oxidative stress via PI3K/Akt/GSK3β and ERK1/2 signaling pathways. Bosn J Basic Med Sci 2021; 21:71-80. [PMID: 31999938 PMCID: PMC7861621 DOI: 10.17305/bjbms.2019.4493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/26/2019] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cell (EPC) transplantation is a safe and effective method to treat acute myocardial infarction (AMI). However, oxidative stress leads to the death of a large number of EPCs in the early stage of transplantation, severely weakening the therapeutic effect. Previous studies demonstrated that microRNAs regulate the biological function of EPCs. The aim of the current study was to investigate the effect of microRNA on the biological function of EPCs under oxidative stress. Quantitative reverse transcription PCR was performed to detect the expression of miR-126, miR-508-5p, miR-150, and miR-16 in EPCs from rats, among which miR-126 showed a relatively higher expression. Treatment with H2O2 decreased miR-126 expression in EPCs in a dose-dependent manner. EPCs were further transfected with miR-126 mimics or inhibitors, followed by H2O2 treatment. Overexpression of miR-126 enhanced the proliferation, migration, and tube formation of H2O2-treated EPCs. MiR-126 overexpression also inhibited reactive oxygen species and malondialdehyde levels and enhanced superoxide dismutase levels, as well as increased angiopoietin (Ang)1 expression and decreased Ang2 expression in H2O2-treated EPCs. Moreover, miR-126 participated in the regulation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in EPCs, where both pathways were activated after miR-126 overexpression in H2O2-treated EPCs. Overall, we showed that miR-126 promoted the biological function of EPCs under H2O2-induced oxidative stress by activating the PI3K/Akt/GSK3β and ERK1/2 signaling pathway, which may serve as a new therapeutic approach to treat AMI.
Collapse
Affiliation(s)
- Qinqin Wu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Benling Qi
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Duan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Ming
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengqin Yan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxia He
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofen Bu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Sun
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Hou L, Yang G, Tang S, Alcazar C, Joshi P, Strassberg Z, Kim M, Kawamura M, Woo YJ, Shrager J, Ding S, Huang NF. Small Molecule Derived From Carboxyethylpyrrole Protein Adducts Promotes Angiogenesis in a Mouse Model of Peripheral Arterial Disease. J Am Heart Assoc 2019; 7:e009234. [PMID: 30371212 PMCID: PMC6222956 DOI: 10.1161/jaha.118.009234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background CEP (ω-[2-carboxyethyl]pyrrole) protein adducts are the end products of lipid oxidation associated with inflammation and have been implicated in the induction of angiogenesis in pathological conditions such as tissue ischemia. We synthesized small molecules derived from CEP protein adducts and evaluated the angiogenic effect of the CEP analog CEP 03 in the setting of peripheral arterial disease. Methods and Results The angiogenic effect of CEP 03 was assessed by in vitro analysis of primary human microvascular endothelial cell proliferation and tubelike formation in Matrigel (Corning). In the presence of CEP 03, proliferation of endothelial cells in vitro increased by 27±18% under hypoxic (1% O2) conditions, reaching similar levels to that of VEGF A (vascular endothelial growth factor A) stimulation (22±10%), relative to the vehicle control treatment. A similar effect of CEP 03 was demonstrated in the increased number of tubelike branches in Matrigel, reaching >70% induction in hypoxia, compared with the vehicle control. The therapeutic potential of CEP 03 was further evaluated in a mouse model of peripheral arterial disease by quantification of blood perfusion recovery and capillary density. In the ischemic hind limb, treatment of CEP 03 encapsulated within Matrigel significantly enhanced blood perfusion by 2-fold after 14 days compared with those treated with Matrigel alone. Moreover, these results concurred with histological finding that treatment of CEP 03 in Matrigel resulted in a significant increase in microvessel density compared with Matrigel alone. Conclusions Our data suggest that CEP 03 has a profound positive effect on angiogenesis and neovessel formation and thus has therapeutic potential for treatment of peripheral arterial disease.
Collapse
Affiliation(s)
- Luqia Hou
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Guang Yang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Shibing Tang
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA
| | - Cynthia Alcazar
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Prajakta Joshi
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Zachary Strassberg
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Michael Kim
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Masashi Kawamura
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Y Joseph Woo
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Joseph Shrager
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Sheng Ding
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA.,5 Department of Pharmaceutical Chemistry University of California San Francisco CA
| | - Ngan F Huang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| |
Collapse
|
8
|
Li X, Lan X, Zhao Y, Wang G, Shi G, Li H, Hu Y, Xu X, Zhang B, Ye K, Gu X, Du C, Wang H. SDF-1/CXCR4 axis enhances the immunomodulation of human endometrial regenerative cells in alleviating experimental colitis. Stem Cell Res Ther 2019; 10:204. [PMID: 31286993 PMCID: PMC6615145 DOI: 10.1186/s13287-019-1298-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/25/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022] Open
Abstract
Endometrial regenerative cells (ERCs) are a new type of mesenchymal-like stromal cells, and their therapeutic potential has been tested in a variety of disease models. SDF-1/CXCR4 axis plays a chemotaxis role in stem/stromal cell migration. The aim of the present study was to investigate the role of SDF-1/CXCR4 axis in the immunomodulation of ERCs on the experimental colitis. The immunomodulation of ERCs in the presence or absence of pretreatment of SDF-1 or AMD3100 was examined in both in vitro cell culture system and dextran sulphate sodium-induced colitis in mice. The results showed that SDF-1 increased the expression of CXCR4 on the surface of ERCs. As compared with normal ERCs, the SDF-1-treated, CXCR4 high-expressing ERCs more significantly suppressed dendritic cell population as well as stimulated both type 2 macrophages and regulatory T cells in vitro and in vivo. Meanwhile, SDF-1-pretreated ERCs increased the generation of anti-inflammatory factors (e.g., IL-4, IL-10) and decreased the pro-inflammatory factors (e.g., IL-6, TNF-α). In addition, SDF-1-pretreated CM-Dil-labeled ERCs were found to engraft to injured colon. Our results may suggest that an SDF-1-induced high level of CXCR4 expression enhances the immunomodulation of ERCs in alleviating experimental colitis in mice.
Collapse
Affiliation(s)
- Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Xu Lan
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiming Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Grace Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ganggang Shi
- Department of Colorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Hongyue Li
- Tianjin General Surgery Institute, Tianjin, China
| | - Yonghao Hu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Xiaoxi Xu
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoren Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Kui Ye
- Department of Vascular Surgery, Tianjin Fourth Central Hospital, Tianjin, China
| | - Xiangying Gu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia, Canada.,Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin General Surgery Institute, Tianjin, China.
| |
Collapse
|
9
|
Gaffey AC, Chen MH, Trubelja A, Venkataraman CM, Chen CW, Chung JJ, Schultz S, Sehgal CM, Burdick JA, Atluri P. Delivery of progenitor cells with injectable shear-thinning hydrogel maintains geometry and normalizes strain to stabilize cardiac function after ischemia. J Thorac Cardiovasc Surg 2018; 157:1479-1490. [PMID: 30579534 DOI: 10.1016/j.jtcvs.2018.07.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The ventricle undergoes adverse remodeling after myocardial infarction, resulting in abnormal biomechanics and decreased function. We hypothesize that tissue-engineered therapy could minimize postischemic remodeling through mechanical stress reduction and retention of tensile myocardial properties due to improved endothelial progenitor cell retention and intrinsic biomechanical properties of the hyaluronic acid shear-thinning gel. METHODS Endothelial progenitor cells were harvested from adult Wistar rats and resuspended in shear-thinning gel. The constructs were injected at the border zone of ischemic rat myocardium in an acute model of myocardial infarction. Myocardial remodeling, tensile properties, and hemodynamic function were analyzed: control (phosphate-buffered saline), endothelial progenitor cells, shear-thinning gel, and shear-thinning gel + endothelial progenitor cells. Novel high-resolution, high-sensitivity ultrasound with speckle tracking allowed for global strain analysis. Uniaxial testing assessed tensile biomechanical properties. RESULTS Shear-thinning gel + endothelial progenitor cell injection significantly increased engraftment and retention of the endothelial progenitor cells within the myocardium compared with endothelial progenitor cells alone. With the use of strain echocardiography, a significant improvement in left ventricular ejection fraction was noted in the shear-thinning gel + endothelial progenitor cell cohort compared with control (69.5% ± 10.8% vs 40.1% ± 4.6%, P = .04). A significant normalization of myocardial longitudinal displacement with subsequent stabilization of myocardial velocity with shear-thinning gel + endothelial progenitor cell therapy compared with control was also evident (0.84 + 0.3 cm/s vs 0.11 ± 0.01 cm/s, P = .03). A significantly positive and higher myocardial strain was observed in shear-thinning gel + endothelial progenitor cell (4.5% ± 0.45%) compared with shear-thinning gel (3.7% ± 0.24%), endothelial progenitor cell (3.5% ± 0.97%), and control (8.6% ± 0.3%, P = .05). A resultant reduction in dynamic stiffness was noted in the shear-thinning gel + endothelial progenitor cell cohort. CONCLUSIONS This novel injectable shear-thinning hyaluronic acid hydrogel demonstrates stabilization of border zone myocardium with reduction in adverse myocardial remodeling and preservation of myocardial biomechanics. The cellular construct provides a normalization of strain measurements and reduces left ventricular dilatation, thus resulting in improvement of left ventricular function.
Collapse
Affiliation(s)
- Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Minna H Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Chantel M Venkataraman
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Carol W Chen
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Jennifer J Chung
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Susan Schultz
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
10
|
Mantakaki A, Fakoya AOJ, Sharifpanah F. Recent advances and challenges on application of tissue engineering for treatment of congenital heart disease. PeerJ 2018; 6:e5805. [PMID: 30386701 PMCID: PMC6204240 DOI: 10.7717/peerj.5805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022] Open
Abstract
Congenital heart disease (CHD) affects a considerable number of children and adults worldwide. This implicates not only developmental disorders, high mortality, and reduced quality of life but also, high costs for the healthcare systems. CHD refers to a variety of heart and vascular malformations which could be very challenging to reconstruct the malformed region surgically, especially when the patient is an infant or a child. Advanced technology and research have offered a better mechanistic insight on the impact of CHD in the heart and vascular system of infants, children, and adults and identified potential therapeutic solutions. Many artificial materials and devices have been used for cardiovascular surgery. Surgeons and the medical industry created and evolved the ball valves to the carbon-based leaflet valves and introduced bioprosthesis as an alternative. However, with research further progressing, contracting tissue has been developed in laboratories and tissue engineering (TE) could represent a revolutionary answer for CHD surgery. Development of engineered tissue for cardiac and aortic reconstruction for developing bodies of infants and children can be very challenging. Nevertheless, using acellular scaffolds, allograft, xenografts, and autografts is already very common. Seeding of cells on surface and within scaffold is a key challenging factor for use of the above. The use of different types of stem cells has been investigated and proven to be suitable for tissue engineering. They are the most promising source of cells for heart reconstruction in a developing body, even for adults. Some stem cell types are more effective than others, with some disadvantages which may be eliminated in the future.
Collapse
Affiliation(s)
| | | | - Fatemeh Sharifpanah
- Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany
| |
Collapse
|
11
|
Hou Y, Li C. Stem/Progenitor Cells and Their Therapeutic Application in Cardiovascular Disease. Front Cell Dev Biol 2018; 6:139. [PMID: 30406100 PMCID: PMC6200850 DOI: 10.3389/fcell.2018.00139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/28/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the world. The stem/progenitor cell-based therapy has emerged as a promising approach for the treatment of a variety of cardiovascular diseases including myocardial infarction, stroke, peripheral arterial disease, and diabetes. An increasing number of evidence has shown that stem/progenitor cell transplantation could replenish damaged cells, improve cardiac and vascular functions, and repair injured tissues in many pre-clinical studies and clinical trials. In this review, we have outlined the major types of stem/progenitor cells, and summarized the studies in applying these cells, especially endothelial stem/progenitor cells and their derivatives, in the treatment of cardiovascular disease. Here the strategies used to improve the stem/progenitor cell-based therapies in cardiovascular disease and the challenges with these therapies in clinical applications are also reviewed.
Collapse
Affiliation(s)
- Yuning Hou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| | - Chunying Li
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
12
|
Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes by Soluble Factors from Human Mesenchymal Stem Cells. Mol Ther 2018; 26:2681-2695. [PMID: 30217728 PMCID: PMC6224789 DOI: 10.1016/j.ymthe.2018.08.012] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/16/2022] Open
Abstract
In this study, we proposed that the functionality or phenotype of differentiated cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs) might be modified by co-culture with mesenchymal stem cells (MSCs), resulting in an improved therapeutic potential for failing myocardial tissues. Structural, motility, electrophysiological, and metabolic analyses revealed that iPSC-CMs co-cultured with MSCs displayed aligned myofibrils with A-, H-, and I-bands that could contract and relax quickly, indicating the promotion of differentiation and the establishment of the iPSC-CM structural framework, and showed clear gap junctions and an electric pacing of >2 Hz, indicating enhanced cell-cell interactions. In addition, soluble factors excreted by MSCs, including several cytokines and exosomes, enhanced cardiomyocyte-specific marker production, produced more energy under normal and stressed conditions, and reduced reactive oxygen species production by iPSC-CMs under stressed condition. Notably, gene ontology and pathway analysis revealed that microRNAs and proteins in the exosomes impacted the functionality and maturation of iPSC-CMs. Furthermore, cell sheets consisting of a mixture of iPSC-CMs and MSCs showed longer survival and enhanced therapeutic effects compared with those consisting of iPSC-CMs alone. This may lead to a new type of iPSC-based cardiomyogenesis therapy for patients with heart failure.
Collapse
|
13
|
Geng X, Liu B, Liu J, Liu D, Lu Y, Sun X, Liang K, Kong B. Interfacial tissue engineering of heart regenerative medicine based on soft cell-porous scaffolds. J Thorac Dis 2018; 10:S2333-S2345. [PMID: 30123574 DOI: 10.21037/jtd.2018.01.117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Myocardial infarction (MI), occurs when the coronary artery is occluded resulting in the hypoxia of areas in heart tissue, is increasing in recent years because of the population ageing and lifestyle changes. Currently, there is no ideal therapeutic scheme because of the limitation of MI therapeutic strategies due to the lack of regenerative ability of the heart cells in adult humans. Recent advances in tissue engineering and regenerative medicine brings hope to the MI therapy and current studies are focusing on restoring the function and structure of damaged tissue by delivering exogenous cells or stimulating endogenous heart cells. However, attempts to directly inject stem cells or cardiomyocytes to the infract zone often lead to rapid cell death and abundant cell loss. To address this challenge, various soft repair cells and porous scaffold materials have been integrated to improve cell retention and engraftment and preventing left ventricle (LV) dilatation. In this article, we will review the current method for heart regeneration based on soft cell-porous scaffold interfacial tissue engineering including common stem cell types, biomaterials, and cardiac patch and will discuss potential future directions in this area.
Collapse
Affiliation(s)
- Xiwen Geng
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, Shanghai 200433, China.,National Supercomputer Research Center of Advanced Materials, Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Bing Liu
- National Supercomputer Research Center of Advanced Materials, Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.,Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250014, China
| | - Jiaqing Liu
- National Supercomputer Research Center of Advanced Materials, Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Dong Liu
- National Supercomputer Research Center of Advanced Materials, Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Yupeng Lu
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250014, China.,School of Materials Science and Engineering, Shandong University, Jinan 250014, China
| | - Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Kang Liang
- School of Chemical Engineering, and Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Biao Kong
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, Shanghai 200433, China
| |
Collapse
|
14
|
Fakoya AOJ, Otohinoyi DA, Yusuf J. Current Trends in Biomaterial Utilization for Cardiopulmonary System Regeneration. Stem Cells Int 2018; 2018:3123961. [PMID: 29853910 PMCID: PMC5949153 DOI: 10.1155/2018/3123961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/15/2017] [Accepted: 03/01/2018] [Indexed: 12/28/2022] Open
Abstract
The cardiopulmonary system is made up of the heart and the lungs, with the core function of one complementing the other. The unimpeded and optimal cycling of blood between these two systems is pivotal to the overall function of the entire human body. Although the function of the cardiopulmonary system appears uncomplicated, the tissues that make up this system are undoubtedly complex. Hence, damage to this system is undesirable as its capacity to self-regenerate is quite limited. The surge in the incidence and prevalence of cardiopulmonary diseases has reached a critical state for a top-notch response as it currently tops the mortality table. Several therapies currently being utilized can only sustain chronically ailing patients for a short period while they are awaiting a possible transplant, which is also not devoid of complications. Regenerative therapeutic techniques now appear to be a potential approach to solve this conundrum posed by these poorly self-regenerating tissues. Stem cell therapy alone appears not to be sufficient to provide the desired tissue regeneration and hence the drive for biomaterials that can support its transplantation and translation, providing not only physical support to seeded cells but also chemical and physiological cues to the cells to facilitate tissue regeneration. The cardiac and pulmonary systems, although literarily seen as just being functionally and spatially cooperative, as shown by their diverse and dissimilar adult cellular and tissue composition has been proven to share some common embryological codevelopment. However, necessitating their consideration for separate review is the immense adult architectural difference in these systems. This review also looks at details on new biological and synthetic biomaterials, tissue engineering, nanotechnology, and organ decellularization for cardiopulmonary regenerative therapies.
Collapse
Affiliation(s)
| | | | - Joshua Yusuf
- All Saints University School of Medicine, Roseau, Dominica
- All Saints University School of Medicine, Kingstown, Saint Vincent and the Grenadines
| |
Collapse
|
15
|
Peng X, Li C, Bai Y, Wang X, Zhang Y, An Y, Teng GJ, Ju S. Noninvasive evaluation of the migration effect of transplanted endothelial progenitor cells in ischemic muscle using a multimodal imaging agent. Int J Nanomedicine 2018; 13:1819-1829. [PMID: 29606873 PMCID: PMC5868615 DOI: 10.2147/ijn.s152976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Endothelial progenitor cells (EPCs) play an important role in repairing ischemia tissues. However, the survival, migration and therapeutic efficacy of EPCs after transplantation need to be better understood for further cell therapy. Purpose This study investigated the migration effect of EPCs labeled with a multimodal imaging agent in a murine ischemic hindlimb model, using magnetic resonance imaging (MRI) and optical imaging after transplantation. Methods EPCs derived from mouse bone marrow were labeled with a multimodal imaging agent and were administered through intracardiac delivery to mice with ischemic hindlimbs. The injected EPCs and their migration effect were observed via MRI and optical imaging in vivo, and then compared to a reference standard based on histological data. The quantification of gadolinium in tissue samples was done using inductively coupled plasma mass spectrometry (ICP-MS). Results Using in vivo MRI and optical imaging, the labeled EPCs were observed to migrate to ischemic muscle on days 3-5 after injection, while ex vivo, the EPCs were observed in the capillary vessels of the injured tissue. There were significant linear correlations between the Gd contents measured using ICP-MS in samples from the ischemic hindlimbs and livers and T1 relaxation times calculated using MRI, as well as the average fluorescence signal intensities recorded in optical images (T1 relaxation time: r=0.491; average signal from optical imaging: r=0.704, P<0.01). EPC treatment upregulated the levels of C-X-C chemokine receptor 4 and vascular endothelial growth factor (VEGF) receptor 2 and enhanced the expression of stromal cell-derived factor-1 and VEGF. Conclusion Transplanted EPCs can be monitored with noninvasive MRI and optical imaging in vivo and were found to enhance the paracrine secretion of angiogenic factors.
Collapse
Affiliation(s)
- Xingui Peng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yingying Bai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Xinyi Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Yanli An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing
| |
Collapse
|
16
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
17
|
Namiri M, Kazemi Ashtiani M, Abbasalizadeh S, Mazidi Z, Mahmoudi E, Nikeghbalian S, Aghdami N, Baharvand H. Improving the biological function of decellularized heart valves through integration of protein tethering and three-dimensional cell seeding in a bioreactor. J Tissue Eng Regen Med 2017; 12:e1865-e1879. [PMID: 29164801 DOI: 10.1002/term.2617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 07/22/2017] [Accepted: 11/09/2017] [Indexed: 12/30/2022]
Abstract
Decellularized xenogeneic heart valves (DHVs) are promising products for valve replacement. However, the widespread clinical application of such products is limited due to the risk of immune reaction, progressive degeneration, inflammation, and calcification. Here, we have developed an optimized decellularization protocol for a xenogeneic heart valve. We improved the biological function of DHVs by protein tethering onto DHV and three-dimensional (3D) cell seeding in a bioreactor. Our results showed that heart valves treated with a Triton X-100 and sodium deoxycholate-based protocol were completely cell-free, with preserved biochemical and biomechanical properties. The immobilization of stromal derived factor-1α (SDF-1α) and basic fibroblast growth factor on DHV significantly improved recellularization with endothelial progenitor cells under the 3D culture condition in the bioreactor compared to static culture conditions. Cell phenotype analysis showed higher fibroblast-like cells and less myofibroblast-like cells in both protein-tethered DHVs. However, SDF-DHV significantly enhanced recellularization both in vitro and in vivo compared to basic fibroblast growth factor DHV and demonstrated less inflammatory cell infiltration. SDF-DHV had less calcification and platelet adhesion. Altogether, integration of SDF-1α immobilization and 3D cell seeding in a bioreactor might provide a novel, promising approach for production of functional heart valves.
Collapse
Affiliation(s)
- Mehrnaz Namiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Mazidi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elena Mahmoudi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saman Nikeghbalian
- Shiraz Transplant Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
18
|
Kawamura M, Paulsen MJ, Goldstone AB, Shudo Y, Wang H, Steele AN, Stapleton LM, Edwards BB, Eskandari A, Truong VN, Jaatinen KJ, Ingason AB, Miyagawa S, Sawa Y, Woo YJ. Tissue-engineered smooth muscle cell and endothelial progenitor cell bi-level cell sheets prevent progression of cardiac dysfunction, microvascular dysfunction, and interstitial fibrosis in a rodent model of type 1 diabetes-induced cardiomyopathy. Cardiovasc Diabetol 2017; 16:142. [PMID: 29096622 PMCID: PMC5668999 DOI: 10.1186/s12933-017-0625-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/24/2017] [Indexed: 12/21/2022] Open
Abstract
Background Diabetes mellitus is a risk factor for coronary artery disease and diabetic cardiomyopathy, and adversely impacts outcomes following coronary artery bypass grafting. Current treatments focus on macro-revascularization and neglect the microvascular disease typical of diabetes mellitus-induced cardiomyopathy (DMCM). We hypothesized that engineered smooth muscle cell (SMC)-endothelial progenitor cell (EPC) bi-level cell sheets could improve ventricular dysfunction in DMCM. Methods Primary mesenchymal stem cells (MSCs) and EPCs were isolated from the bone marrow of Wistar rats, and MSCs were differentiated into SMCs by culturing on a fibronectin-coated dish. SMCs topped with EPCs were detached from a temperature-responsive culture dish to create an SMC-EPC bi-level cell sheet. A DMCM model was induced by intraperitoneal streptozotocin injection. Four weeks after induction, rats were randomized into 3 groups: control (no DMCM induction), untreated DMCM, and treated DMCM (cell sheet transplant covering the anterior surface of the left ventricle). Results SMC-EPC cell sheet therapy preserved cardiac function and halted adverse ventricular remodeling, as demonstrated by echocardiography and cardiac magnetic resonance imaging at 8 weeks after DMCM induction. Myocardial contrast echocardiography demonstrated that myocardial perfusion and microvascular function were preserved in the treatment group compared with untreated animals. Histological analysis demonstrated decreased interstitial fibrosis and increased microvascular density in the SMC-EPC cell sheet-treated group. Conclusions Treatment of DMCM with tissue-engineered SMC-EPC bi-level cell sheets prevented cardiac dysfunction and microvascular disease associated with DMCM. This multi-lineage cellular therapy is a novel, translatable approach to improve microvascular disease and prevent heart failure in diabetic patients.
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA.,Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Japan
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA.,Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Japan
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Lyndsay M Stapleton
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Bryan B Edwards
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Vi N Truong
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Kevin J Jaatinen
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Arnar B Ingason
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Japan
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
19
|
Li Q, Xia S, Fang H, Pan J, Jia Y, Deng G. VEGF treatment promotes bone marrow-derived CXCR4 + mesenchymal stromal stem cell differentiation into vessel endothelial cells. Exp Ther Med 2017; 13:449-454. [PMID: 28352314 PMCID: PMC5348687 DOI: 10.3892/etm.2017.4019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/11/2016] [Indexed: 01/08/2023] Open
Abstract
Stem/progenitor cells serve an important role in the process of blood vessel repair. However, the mechanism of vascular repair mediated by C-X-C chemokine receptor type 4-positive (CXCR4+) bone marrow-derived mesenchymal stem cells (BMSCs) following myocardial infarction remains unclear. The aim of the present study was to investigate the effects of vascular endothelial growth factor (VEGF) on vessel endothelial differentiation from BMSCs. CXCR4+ BMSCs were isolated from the femoral bone marrow of 2-month-old mice and the cells were treated with VEGF. Expression of endothelial cell markers and the functional properties were assessed by reverse transcription-quantitative polymerase chain reaction, flow cytometry and vascular formation analyses. The results indicated that the CXCR4+ BMSCs from femoral bone marrow cells expressed putative cell surface markers of mesenchymal stem cells. Treatment with VEGF induced platelet/endothelial cell adhesion molecule-1 (PECAM-1) and von Willebrand factor (vWF) expression at the transcriptional and translational levels, compared with untreated controls. Moreover, VEGF treatment induced CXCR4+ BMSCs to form hollow tube-like structures on Matrigel, suggesting that the differentiated endothelial cells had the functional properties of blood vessels. The results demonstrate that the CXCR4+ BMSCs were able to differentiate into vessel endothelial cells following VEGF treatment. For cell transplantation in vascular disease, it may be concluded that CXCR4+ BMSCs are a novel source of endothelial progenitor cells with high potential for application in vascular repair.
Collapse
Affiliation(s)
- Qiming Li
- Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Shudong Xia
- Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Hanyun Fang
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Jiansheng Pan
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Yinfeng Jia
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Gang Deng
- The Ningbo Central Blood Station, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
20
|
Li Z, Shen Z, Du L, He J, Chen S, Zhang J, Luan Y, Fu G. Fn14 is regulated via the RhoA pathway and mediates nuclear factor-kappaB activation by Angiotensin II. Am J Transl Res 2016; 8:5386-5398. [PMID: 28078010 PMCID: PMC5209490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/06/2016] [Indexed: 06/06/2023]
Abstract
Angiotesin II (Ang II) plays an important role in cardiac remodeling. Fibroblast growth factor inducible-14 (Fn14) is the smallest member of the tumor necrosis factor superfamily of receptors. Currently, little is known about the functional role of Fn14 in the heart. Chiefly, we observe the up-regulation of extracellular matrix in in vivo model. We therefore assess the expression and regulation of Fn14 in cardiomyocytes and in vivo models induced by Ang II. In order to study the regulation of Fn14, cardiac remodeling was established in rats and neonatal cardiomyocytes were used in in vitro model. As well, Ang II is able to strongly induce Fn14 expression in in vivo and in vitro models. Fn14 is mediated via RhoA pathways, since siRNA against RhoA prevented the expression of Fn14 in cardiomyocytes. Pretreatment of cardiomyoctes with siRNA against NF-κB and IκBα also decreased Fn14 expression induced by Ang II. We here describe for the first time Ang II regulation of Fn14 in in vivo and in vitro models via RhoA, NF-κB and NF-κB driven gene signaling pathway. In conclusion, Fn14 may be important in regulating the process of cardiac remodeling induced by Ang II.
Collapse
Affiliation(s)
- Zhengwei Li
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Lailing Du
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Jialin He
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Shengyu Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Jiefang Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| |
Collapse
|
21
|
Kang J, Kim TW, Hur J, Kim HS. Strategy to Prime the Host and Cells to Augment Therapeutic Efficacy of Progenitor Cells for Patients with Myocardial Infarction. Front Cardiovasc Med 2016; 3:46. [PMID: 27933299 PMCID: PMC5121226 DOI: 10.3389/fcvm.2016.00046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/08/2016] [Indexed: 11/23/2022] Open
Abstract
Cell therapy in myocardial infarction (MI) is an innovative strategy that is regarded as a rescue therapy to repair the damaged myocardium and to promote neovascularization for the ischemic border zone. Among several stem cell sources for this purpose, autologous progenitors from bone marrow or peripheral blood would be the most feasible and safest cell-source. Despite the theoretical benefit of cell therapy, this method is not widely adopted in the actual clinical practice due to its low therapeutic efficacy. Various methods have been used to augment the efficacy of cell therapy in MI, such as using different source of progenitors, genetic manipulation of cells, or priming of the cells or hosts (patients) with agents. Among these methods, the strategy to augment the therapeutic efficacy of the autologous peripheral blood mononuclear cells (PBMCs) by priming agents may be the most feasible and the safest method that can be applied directly to the clinic. In this review, we will discuss the current status and future directions of priming PBMCs or patients, as for cell therapy of MI.
Collapse
Affiliation(s)
- Jeehoon Kang
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea
| | - Tae-Won Kim
- Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Jin Hur
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital , Seoul , South Korea
| | - Hyo-Soo Kim
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
22
|
Feyen DA, Gaetani R, Doevendans PA, Sluijter JP. Stem cell-based therapy: Improving myocardial cell delivery. Adv Drug Deliv Rev 2016; 106:104-115. [PMID: 27133386 DOI: 10.1016/j.addr.2016.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022]
Abstract
Stem cell-based therapies form an exciting new class of medicine that attempt to provide the body with the building blocks required for the reconstruction of damaged organs. However, delivering cells to the correct location, while preserving their integrity and functional properties, is a complex undertaking. These challenges have led to the development of a highly dynamic interdisciplinary research field, wherein medical, biological, and chemical sciences have collaborated to develop strategies to overcome the physiological barriers imposed on the cellular therapeutics. In this respect, improving the acute retention and subsequent survival of stem cells is key to effectively increase the effect of the therapy, while proper tissue integration is imperative for stem cells to functionally replace lost cells in damaged organs. In this review, we will use the heart as an example to highlight the current knowledge of therapeutic stem cell utilization, the existing pitfalls and limitations, and the approaches that have been developed to overcome them.
Collapse
|
23
|
Kim JJ, Hou L, Huang NF. Vascularization of three-dimensional engineered tissues for regenerative medicine applications. Acta Biomater 2016; 41:17-26. [PMID: 27262741 PMCID: PMC4969172 DOI: 10.1016/j.actbio.2016.06.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 01/05/2023]
Abstract
UNLABELLED Engineering of three-dimensional (3D) tissues is a promising approach for restoring diseased or dysfunctional myocardium with a functional replacement. However, a major bottleneck in this field is the lack of efficient vascularization strategies, because tissue constructs produced in vitro require a constant flow of oxygen and nutrients to maintain viability and functionality. Compared to angiogenic cell therapy and growth factor treatment, bioengineering approaches such as spatial micropatterning, integration of sacrificial materials, tissue decellularization, and 3D bioprinting enable the generation of more precisely controllable neovessel formation. In this review, we summarize the state-of-the-art approaches to develop 3D tissue engineered constructs with vasculature, and demonstrate how some of these techniques have been applied towards regenerative medicine for treatment of heart failure. STATEMENT OF SIGNIFICANCE Tissue engineering is a promising approach to replace or restore dysfunctional tissues/organs, but a major bottleneck in realizing its potential is the challenge of creating scalable 3D tissues. Since most 3D engineered tissues require a constant supply of nutrients, it is necessary to integrate functional vasculature within the tissues in order to facilitate the transport of nutrients. To address these needs, researchers are employing biomaterial engineering and design strategies to foster vessel formation within 3D tissues. This review highlights the state-of-the-art bioengineering tools and technologies to create vascularized 3D tissues for clinical applications in regenerative medicine, highlighting the application of these technologies to engineer vascularized cardiac patches for treatment of heart failure.
Collapse
Affiliation(s)
- Joseph J Kim
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Luqia Hou
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, 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 University, Stanford, CA, USA.
| |
Collapse
|
24
|
Overexpression of protein kinase C ɛ improves retention and survival of transplanted mesenchymal stem cells in rat acute myocardial infarction. Cell Death Dis 2016; 7:e2056. [PMID: 26775707 PMCID: PMC4816190 DOI: 10.1038/cddis.2015.417] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 12/13/2015] [Accepted: 12/15/2015] [Indexed: 01/09/2023]
Abstract
We assessed the effects of protein kinase C ɛ (PKCɛ) for improving stem cell therapy for acute myocardial infarction (AMI). Primary mesenchymal stem cells (MSCs) were harvested from rat bone marrow. PKCɛ-overexpressed MSCs and control MSCs were transplanted into infarct border zones in a rat AMI model. MSCs and PKCɛ distribution and expression of principal proteins involved in PKCɛ signaling through the stromal cell-derived factor 1 (SDF-1)/CXC chemokine receptor type 4 (CXCR4) axis and the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) pathway were analyzed by immunofluorescence and western blot 1 day after transplantation. Echocardiographic measurements and histologic studies were performed at 4 weeks after transplantation, and MSC survival, expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), transforming growth factor β (TGFβ), cardiac troponin I (cTnI), von Willebrand factor (vWF), smooth muscle actin (SMA) and factor VIII and apoptosis in infarct border zones were assessed. Rat heart muscles retained more MSCs and SDF-1, CXCR4, PI3K and phosphorylated AKT increased with PKCɛ overexpression 1 day after transplantation. MSC survival and VEGF, bFGF, TGFβ, cTnI, vWF, SMA and factor VIII expression increased in animals with PKCɛ-overexpressed MSCs at 4 weeks after transplantation and cardiac dysfunction and remodeling improved. Infarct size and apoptosis decreased as well. Inhibitory actions of CXCR4 or PI3K partly attenuated the effects of PKCɛ. Activation of PKCɛ may improve retention, survival and differentiation of transplanted MSCs in myocardia. Augmentation of PKCɛ expression may enhance the therapeutic effects of stem cell therapy for AMI.
Collapse
|
25
|
Gaffey AC, Chen MH, Venkataraman CM, Trubelja A, Rodell CB, Dinh PV, Hung G, MacArthur JW, Soopan RV, Burdick JA, Atluri P. Injectable shear-thinning hydrogels used to deliver endothelial progenitor cells, enhance cell engraftment, and improve ischemic myocardium. J Thorac Cardiovasc Surg 2015; 150:1268-76. [PMID: 26293548 PMCID: PMC4637242 DOI: 10.1016/j.jtcvs.2015.07.035] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/30/2015] [Accepted: 07/12/2015] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The clinical translation of cell-based therapies for ischemic heart disease has been limited because of low cell retention (<1%) within, and poor targeting to, ischemic myocardium. To address these issues, we developed an injectable hyaluronic acid (HA) shear-thinning hydrogel (STG) and endothelial progenitor cell (EPC) construct (STG-EPC). The STG assembles as a result of interactions of adamantine- and β-cyclodextrin-modified HA. It is shear-thinning to permit delivery via a syringe, and self-heals upon injection within the ischemic myocardium. This directed therapy to the ischemic myocardial border zone enables direct cell delivery to address adverse remodeling after myocardial infarction. We hypothesize that this system will enhance vasculogenesis to improve myocardial stabilization in the context of a clinically translatable therapy. METHODS Endothelial progenitor cells (DiLDL(+) VEGFR2(+) CD34(+)) were harvested from adult male rats, cultured, and suspended in the STG. In vitro viability was quantified using a live-dead stain of EPCs. The STG-EPC constructs were injected at the border zone of ischemic rat myocardium after acute myocardial infarction (left anterior descending coronary artery ligation). The migration of the enhanced green fluorescent proteins from the construct to ischemic myocardium was analyzed using fluorescent microscopy. Vasculogenesis, myocardial remodeling, and hemodynamic function were analyzed in 4 groups: control (phosphate buffered saline injection); intramyocardial injection of EPCs alone; injection of the STG alone; and treatment with the STG-EPC construct. Hemodynamics and ventricular geometry were quantified using echocardiography and Doppler flow analysis. RESULTS Endothelial progenitor cells demonstrated viability within the STG. A marked increase in EPC engraftment was observed 1-week postinjection within the treated myocardium with gel delivery, compared with EPC injection alone (17.2 ± 0.8 cells per high power field (HPF) vs 3.5 cells ± 1.3 cells per HPF, P = .0002). A statistically significant increase in vasculogenesis was noted with the STG-EPC construct (15.3 ± 5.8 vessels per HPF), compared with the control (P < .0001), EPC (P < .0001), and STG (P < .0001) groups. Statistically significant improvements in ventricular function, scar fraction, and geometry were noted after STG-EPC treatment compared with the control. CONCLUSIONS A novel injectable shear-thinning HA hydrogel seeded with EPCs enhanced cell retention and vasculogenesis after delivery to ischemic myocardium. This therapy limited adverse myocardial remodeling while preserving contractility.
Collapse
Affiliation(s)
- Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Minna H Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Chantel M Venkataraman
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | | | - Patrick V Dinh
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - George Hung
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - John W MacArthur
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Renganaden V Soopan
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
26
|
Shudo Y, Cohen JE, MacArthur JW, Goldstone AB, Otsuru S, Trubelja A, Patel J, Edwards BB, Hung G, Fairman AS, Brusalis C, Hiesinger W, Atluri P, Hiraoka A, Miyagawa S, Sawa Y, Woo YJ. A Tissue-Engineered Chondrocyte Cell Sheet Induces Extracellular Matrix Modification to Enhance Ventricular Biomechanics and Attenuate Myocardial Stiffness in Ischemic Cardiomyopathy. Tissue Eng Part A 2015; 21:2515-25. [PMID: 26154752 DOI: 10.1089/ten.tea.2014.0155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There exists a substantial body of work describing cardiac support devices to mechanically support the left ventricle (LV); however, these devices lack biological effects. To remedy this, we implemented a cell sheet engineering approach utilizing chondrocytes, which in their natural environment produce a relatively elastic extracellular matrix (ECM) for a cushioning effect. Therefore, we hypothesized that a chondrocyte cell sheet applied to infarcted and borderzone myocardium will biologically enhance the ventricular ECM and increase elasticity to augment cardiac function in a model of ischemic cardiomyopathy (ICM). Primary articular cartilage chondrocytes of Wistar rats were isolated and cultured on temperature-responsive culture dishes to generate cell sheets. A rodent ICM model was created by ligating the left anterior descending coronary artery. Rats were divided into two groups: cell sheet transplantation (1.0 × 10(7) cells/dish) and no treatment. The cell sheet was placed onto the surface of the heart covering the infarct and borderzone areas. At 4 weeks following treatment, the decreased fibrotic extension and increased elastic microfiber networks in the infarct and borderzone areas correlated with this technology's potential to stimulate ECM formation. The enhanced ventricular elasticity was further confirmed by the axial stretch test, which revealed that the cell sheet tended to attenuate tensile modulus, a parameter of stiffness. This translated to increased wall thickness in the infarct area, decreased LV volume, wall stress, mass, and improvement of LV function. Thus, the chondrocyte cell sheet strengthens the ventricular biomechanical properties by inducing the formation of elastic microfiber networks in ICM, resulting in attenuated myocardial stiffness and improved myocardial function.
Collapse
Affiliation(s)
- Yasuhiro Shudo
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
- 4 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Jeffrey E Cohen
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - John W MacArthur
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Andrew B Goldstone
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Satoru Otsuru
- 3 Center for Childhood Cancer and Blood Diseases, The Research Institute , Nationwide Children's Hospital, Columbus, Ohio
| | - Alen Trubelja
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Jay Patel
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
| | - Bryan B Edwards
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
| | - George Hung
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Alexander S Fairman
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Christopher Brusalis
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - William Hiesinger
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Pavan Atluri
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Arudo Hiraoka
- 2 Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Shigeru Miyagawa
- 4 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Yoshiki Sawa
- 4 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Y Joseph Woo
- 1 Department of Cardiothoracic Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
27
|
Tada-Oikawa S, Ichihara G, Suzuki Y, Izuoka K, Wu W, Yamada Y, Mishima T, Ichihara S. Zn(II) released from zinc oxide nano/micro particles suppresses vasculogenesis in human endothelial colony-forming cells. Toxicol Rep 2015; 2:692-701. [PMID: 28962405 PMCID: PMC5598154 DOI: 10.1016/j.toxrep.2015.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 11/30/2022] Open
Abstract
Zinc oxide (ZnO) nanoparticles have been widely used in industry, cosmetics, and biomedicine. Recent studies suggested that these nanoparticles could have a major impact on the cardiovascular system. Endothelial progenitor cells (EPCs) contribute to postnatal endothelial repair and regeneration. The present study dissected the effects of ZnO nanoparticles on vasculogenesis using human endothelial colony forming cells (ECFCs), which participate in post-natal vasculogenesis. Two types of ZnO particles were used (nano and micro), in addition to zinc chloride solutions with zinc ion concentrations equal to those in ZnO nanoparticles. Twenty-four-hour exposure induced cytotoxicity in a dose-dependent manner and increased ECFCs apoptosis in all groups. The exposure also reduced the functional capacity of ECFCs on Matrix gel to form tubules, compared with the control cells. These effects were associated with downregulation of expression of vascular endothelial growth factor receptor, VEGFR2 and CXC chemokine receptor, CXCR4. The results suggest that ZnO nanoparticles suppress vasculogenesis from ECFCs through downregulation of the expression of receptors related to vasculogenesis. These effects are based the concentration of released Zn(II).
Collapse
Affiliation(s)
- Saeko Tada-Oikawa
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo Univeristy of Science, Noda, Japan
| | - Yuka Suzuki
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Kiyora Izuoka
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Wenting Wu
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiji Yamada
- Life Science Research Center, Mie University, Tsu, Japan
| | - Takashi Mishima
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
- Life Science Research Center, Mie University, Tsu, Japan
| |
Collapse
|
28
|
He H, Zhao ZH, Han FS, Wang XF, Zeng YJ. Activation of protein kinase C ε enhanced movement ability and paracrine function of rat bone marrow mesenchymal stem cells partly at least independent of SDF-1/CXCR4 axis and PI3K/AKT pathway. Int J Clin Exp Med 2015; 8:188-202. [PMID: 25784988 PMCID: PMC4358443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/07/2015] [Indexed: 06/04/2023]
Abstract
OBJECTS to probe into the effects of PKCε on migration and paracrine functions of stem cells and potential molecular mechanisms. METHODS Bone Marrow mesenchymal stem cells (BMMSCs) were obtained from rat femur and passaged. mRNA and protein levels of capital proteins in PKCε signaling, SDF-1/CXCR4 axis and PI3K/AKT pathway in the MSCs in different conditions treating with PKC agonist, specific PKCε inhibitor, CXCR4 antagonist or PI3K inhibitor for 24 hours were analyzed by real-time PCR and western blot, and migration abilities were observed by migration assay in vitro and the changes of paracrine factors in different treatments were analyzed by protein clips assay. RESULTS the levels of p-JNK, p-P38MAPK, SDF-1, CXCR4, PI3K and p-AKT increased significantly after treating with PKC agonist (P < 0.05) and decreased obviously after treating with specific PKCε inhibitor. Migration ability and paracrine function of MSCs were enhanced in PMA group and attenuated in PKCε inhibitor group, and inhibiting activity of CXCR4 or PI3K attenuated the effects of PKCε, but not abolished completely. CONCLUSION There was cross-talking between PKCε signaling and SDF-1/CXCR4 axis and PI3K/AKT pathway in signal transduction of MSCs. Activating PKCε could improve migration ability and paracrine function of MSCs partly at least independent of SDF-1/CXCR4 axis and PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hua He
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical UniversityBeijing 100029, China
| | - Zhi-Hong Zhao
- Department of Cardiology, Pudong New Area District Zhoupu HospitalShanghai 201318, China
| | - Fu-Sheng Han
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical UniversityBeijing 100029, China
| | - Xi-Fu Wang
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical UniversityBeijing 100029, China
| | - Yu-Jie Zeng
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical UniversityBeijing 100029, China
| |
Collapse
|
29
|
Ban K, Park HJ, Kim S, Andukuri A, Cho KW, Hwang JW, Cha HJ, Kim SY, Kim WS, Jun HW, Yoon YS. Cell therapy with embryonic stem cell-derived cardiomyocytes encapsulated in injectable nanomatrix gel enhances cell engraftment and promotes cardiac repair. ACS NANO 2014; 8:10815-25. [PMID: 25210842 PMCID: PMC4212793 DOI: 10.1021/nn504617g] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/11/2014] [Indexed: 05/25/2023]
Abstract
A significant barrier to the therapeutic use of stem cells is poor cell retention in vivo. Here, we evaluate the therapeutic potential and long-term engraftment of cardiomyocytes (CMs) derived from mouse embryonic stem cells (mESCs) encapsulated in an injectable nanomatrix gel consisting of peptide amphiphiles incorporating cell adhesive ligand Arg-Gly-Asp-Ser (PA-RGDS) in experimental myocardial infarction (MI). We cultured rat neonatal CMs in PA-RGDS for 7 days and found that more than 90% of the CMs survived. Next, we intramyocardially injected mouse CM cell line HL-1 CMs with or without PA-RGDS into uninjured hearts. Histologic examination and flow cytometry analysis of digested heart tissues showed approximately 3-fold higher engraftment in the mice that received CMs with PA-RGDS compared to those without PA-RGDS. We further investigated the therapeutic effects and long-term engraftment of mESC-CMs with PA-RGDS on MI in comparison with PBS control, CM-only, and PA-RGDS only. Echocardiography demonstrated that the CM-only and CM+PA-RGDS groups showed higher cardiac function at week 2 compared to other groups. However, from 3 weeks, higher cardiac function was maintained only in the CM+PA-RGDS group; this was sustained for 12 weeks. Confocal microscopic examination of the cardiac tissues harvested at 14 weeks demonstrated sustained engraftment and integration of mESC-CMs into host myocardium in the CM+PA-RGDS group only. This study for the first time demonstrated that PA-RGDS encapsulation can enhance survival of mESC-derived CMs and improve cardiac function post-MI. This nanomatrix gel-mediated stem cell therapy can be a promising option for treating MI.
Collapse
Affiliation(s)
- Kiwon Ban
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Hun-Jun Park
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sangsung Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Adinarayana Andukuri
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Kyu-Won Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jung Wook Hwang
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Ho Jin Cha
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Sang Yoon Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Woan-Sang Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama 35203, United States
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
30
|
Yuan X, He B, Lv Z, Luo S. Fabrication of self-assembling peptide nanofiber hydrogels for myocardial repair. RSC Adv 2014. [DOI: 10.1039/c4ra08582e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
31
|
Loss of Sirt3 limits bone marrow cell-mediated angiogenesis and cardiac repair in post-myocardial infarction. PLoS One 2014; 9:e107011. [PMID: 25192254 PMCID: PMC4156371 DOI: 10.1371/journal.pone.0107011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/07/2014] [Indexed: 12/22/2022] Open
Abstract
Sirtuin-3 (Sirt3) has a critical role in the regulation of human aging and reactive oxygen species (ROS) formation. A recent study has identified Sirt3 as an essential regulator of stem cell aging. This study investigated whether Sirt3 is necessary for bone marrow cell (BMC)-mediated cardiac repair in post-myocardial infarction (MI). In vitro, BMC-derived endothelial progenitor cells (EPCs) from wild type (WT) and Sirt3KO mice were cultured. EPC angiogenesis, ROS formation and apoptosis were assessed. In vivo, WT and Sirt3 KO mice were subjected to MI and BMCs from WT and Sirt3 KO mice were injected into ischemic area immediately. The expression of VEGF and VEGFR2 was reduced in Sirt3KO-EPCs. Angiogenic capacities and colony formation were significantly impaired in Sirt3KO-EPCs compared to WT-EPCs. Loss of Sirt3 further enhanced ROS formation and apoptosis in EPCs. Overexpression of Sirt3 or treatment with NADPH oxidase inhibitor apocynin (Apo, 200 and 400 microM) rescued these abnormalities. In post-MI mice, BMC treatment increased number of Sca1+/c-kit+ cells; enhanced VEGF expression and angiogenesis whereas Sirt3KO-BMC treatment had little effects. BMC treatment also attenuated NADPH oxidase subunits p47phox and gp91phox expression, and significantly reduced ROS formation, apoptosis, fibrosis and hypertrophy in post-MI mice. Sirt3KO-BMC treatment did not display these beneficial effects. In contrast, Sirt3KO mice treated with BMCs from WT mice attenuated myocardial apoptosis, fibrosis and improved cardiac function. Our data demonstrate that Sirt3 is essential for BMC therapy; and loss of Sirt3 limits BMC-mediated angiogenesis and cardiac repair in post-MI.
Collapse
|
32
|
Pereira CL, Gonçalves RM, Peroglio M, Pattappa G, D'Este M, Eglin D, Barbosa MA, Alini M, Grad S. The effect of hyaluronan-based delivery of stromal cell-derived factor-1 on the recruitment of MSCs in degenerating intervertebral discs. Biomaterials 2014; 35:8144-53. [DOI: 10.1016/j.biomaterials.2014.06.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/06/2014] [Indexed: 12/15/2022]
|
33
|
Hung HS, Yang YC, Lin YC, Lin SZ, Kao WC, Hsieh HH, Chu MY, Fu RH, Hsu SH. Regulation of human endothelial progenitor cell maturation by polyurethane nanocomposites. Biomaterials 2014; 35:6810-21. [DOI: 10.1016/j.biomaterials.2014.04.076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/22/2014] [Indexed: 12/24/2022]
|
34
|
Atluri P, Miller JS, Emery RJ, Hung G, Trubelja A, Cohen JE, Lloyd K, Han J, Gaffey AC, MacArthur JW, Chen CS, Woo YJ. Tissue-engineered, hydrogel-based endothelial progenitor cell therapy robustly revascularizes ischemic myocardium and preserves ventricular function. J Thorac Cardiovasc Surg 2014; 148:1090-7; discussion 1097-8. [PMID: 25129603 DOI: 10.1016/j.jtcvs.2014.06.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Cell-based angiogenic therapy for ischemic heart failure has had limited clinical impact, likely related to low cell retention (<1%) and dispersion. We developed a novel, tissue-engineered, hydrogel-based cell-delivery strategy to overcome these limitations and provide prolonged regional retention of myocardial endothelial progenitor cells at high cell dosage. METHODS Endothelial progenitor cells were isolated from Wistar rats and encapsulated in fibrin gels. In vitro viability was quantified using a fluorescent live-dead stain of transgenic enhanced green fluorescent protein(+) endothelial progenitor cells. Endothelial progenitor cell-laden constructs were implanted onto ischemic rat myocardium in a model of acute myocardial infarction (left anterior descending ligation) for 4 weeks. Intramyocardial cell injection (2 × 10(6) endothelial progenitor cells), empty fibrin, and isolated left anterior descending ligation groups served as controls. Hemodynamics were quantified using echocardiography, Doppler flow analysis, and intraventricular pressure-volume analysis. Vasculogenesis and ventricular geometry were quantified. Endothelial progenitor cell migration was analyzed by using endothelial progenitor cells from transgenic enhanced green fluorescent protein(+) rodents. RESULTS Endothelial progenitor cells demonstrated an overall 88.7% viability for all matrix and cell conditions investigated after 48 hours. Histologic assessment of 1-week implants demonstrated significant migration of transgenic enhanced green fluorescent protein(+) endothelial progenitor cells from the fibrin matrix to the infarcted myocardium compared with intramyocardial cell injection (28 ± 12.3 cells/high power field vs 2.4 ± 2.1 cells/high power field, P = .0001). We also observed a marked increase in vasculogenesis at the implant site. Significant improvements in ventricular hemodynamics and geometry were present after endothelial progenitor cell-hydrogel therapy compared with control. CONCLUSIONS We present a tissue-engineered, hydrogel-based endothelial progenitor cell-mediated therapy to enhance cell delivery, cell retention, vasculogenesis, and preservation of myocardial structure and function.
Collapse
Affiliation(s)
- Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | | | - Robert J Emery
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - George Hung
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jeffrey E Cohen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Kelsey Lloyd
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jason Han
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - John W MacArthur
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | | | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif.
| |
Collapse
|
35
|
The role of integrin α2 in cell and matrix therapy that improves perfusion, viability and function of infarcted myocardium. Biomaterials 2014; 35:4749-58. [PMID: 24631247 DOI: 10.1016/j.biomaterials.2014.02.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/16/2014] [Indexed: 12/13/2022]
Abstract
Injectable delivery matrices hold promise in enhancing engraftment and the overall efficacy of cardiac cell therapies; however, the mechanisms responsible remain largely unknown. Here we studied the interaction of a collagen matrix with circulating angiogenic cells (CACs) in a mouse myocardial infarction model. CACs + matrix treatment enhanced CAC engraftment, and improved myocardial perfusion, viability and function compared to cells or matrix alone. Integrin-linked kinase (ILK) was up-regulated in matrix-cultured CACs. Integrin α2β1 blocking prevented ILK up-regulation, significantly reduced the adhesion, proliferation, and paracrine properties of matrix-cultured CACs, and negated the benefits of CACs + matrix therapy in vivo. Furthermore, integrin α5 was essential for the angiogenic potential of CACs on matrix. These findings indicate that the synergistic therapeutic effect of CACs + matrix therapy in MI requires the matrix to enhance CAC function via α2β1 and α5 integrin signaling mechanisms, rather than simply delivering the cells.
Collapse
|
36
|
A modified approach to induce predictable congestive heart failure by volume overload in rats. PLoS One 2014; 9:e87531. [PMID: 24498127 PMCID: PMC3909118 DOI: 10.1371/journal.pone.0087531] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 12/28/2013] [Indexed: 01/15/2023] Open
Abstract
The model of infrarenal aortocaval fistula (ACF) has recently gained new interest in its use to investigate cardiac pathophysiology. Since in previous investigations the development of congestive heart failure (CHF) was inconsistent and started to develop earliest 8–10 weeks after fistula induction using a 18G needle, this project aimed to induce a predictable degree of CHF within a definite time period using a modified approach. An aortocaval fistula was induced in male Wistar rats using a 16G needle as a modification of the former 18G needle-technique described by Garcia and Diebold. Results revealed within 28±2 days of ACF significantly increased heart and lung weight indices in the ACF group accompanied by elevated filling pressure. All hemodynamic parameters derived from a pressure-volume conductance-catheter in vivo were significantly altered in the ACF consistent with severe systolic and diastolic left ventricular dysfunction. This was accompanied by systemic neurohumoral activation as demonstrated by elevated rBNP-45 plasma concentrations in every rat of the ACF group. Furthermore, the restriction in overall cardiac function was associated with a β1- and β2-adrenoreceptor mRNA downregulation in the left ventricle. In contrast, β3-adrenoreceptor mRNA was upregulated. Finally, electron microscopy of the left ventricle of rats in the ACF group showed signs of progressive subcellular myocardial fragmentation. In conclusion, the morphometric, hemodynamic and neurohumoral characterization of the modified approach revealed predictable and consistent signs of congestive heart failure within 28±2 days. Therefore, this modified approach might facilitate the examination of various questions specific to CHF and allow for pharmacological interventions to determine pathophysiological pathways.
Collapse
|
37
|
Shudo Y, Miyagawa S, Ohkura H, Fukushima S, Saito A, Shiozaki M, Kawaguchi N, Matsuura N, Shimizu T, Okano T, Matsuyama A, Sawa Y. Addition of mesenchymal stem cells enhances the therapeutic effects of skeletal myoblast cell-sheet transplantation in a rat ischemic cardiomyopathy model. Tissue Eng Part A 2014; 20:728-39. [PMID: 24164292 DOI: 10.1089/ten.tea.2012.0534] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Functional skeletal myoblasts (SMBs) are transplanted into the heart effectively and safely as cell sheets, which induce functional recovery in myocardial infarction (MI) patients without lethal arrhythmia. However, their therapeutic effect is limited by ischemia. Mesenchymal stem cells (MSCs) have prosurvival/proliferation and antiapoptotic effects on co-cultured cells in vitro. We hypothesized that adding MSCs to the SMB cell sheets might enhance SMB survival post-transplantation and improve their therapeutic effects. METHODS AND RESULTS Cell sheets of primary SMBs of male Lewis rats (r-SMBs), primary MSCs of human female fat tissues (h-MSCs), and their co-cultures were generated using temperature-responsive dishes. The levels of candidate paracrine factors, rat hepatocyte growth factor and vascular endothelial growth factor, in vitro were significantly greater in the h-MSC/r-SMB co-cultures than in those containing r-SMBs only, by real-time PCR and enzyme-linked immunosorbent assay (ELISA). MI was generated by left-coronary artery occlusion in female athymic nude rats. Two weeks later, co-cultured r-SMB or h-MSC cell sheets were implanted or no treatment was performed (n=10 each). Eight weeks later, systolic and diastolic function parameters were improved in all three treatment groups compared to no treatment, with the greatest improvement in the co-cultured cell sheet transplantation group. Consistent results were found for capillary density, collagen accumulation, myocyte hypertrophy, Akt-signaling, STAT3 signaling, and survival of transplanted cells of rat origin, and were related to poly (ADP-ribose) polymerase-dependent signal transduction. CONCLUSIONS Adding MSCs to SMB cell sheets enhanced the sheets' angiogenesis-related paracrine mechanics and, consequently, functional recovery in a rat MI model, suggesting a possible strategy for clinical applications.
Collapse
Affiliation(s)
- Yasuhiro Shudo
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tong G, Wang N, Zhou Y, Leng J, Gao W, Tong X, Shen Y, Yang J, Ye X, Zhou L, Gao Y. Role of Stromal Cell-Derived Factor-1 in Patients With Non-ST Elevation Acute Coronary Syndrome. Int Heart J 2014; 55:219-27. [DOI: 10.1536/ihj.13-289] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Guoxin Tong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Ningfu Wang
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Jianhang Leng
- Department of Clinical Laboratory Medicine, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Wei Gao
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Xiaoshan Tong
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Yun Shen
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Jianmin Yang
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Xianhua Ye
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Liang Zhou
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| | - Yan Gao
- Department of Cardiology, Hangzhou Hospital (Hangzhou No.1 Municipal Hospital), Nanjing Medical University
| |
Collapse
|
39
|
Macarthur JW, Cohen JE, McGarvey JR, Shudo Y, Patel JB, Trubelja A, Fairman AS, Edwards BB, Hung G, Hiesinger W, Goldstone AB, Atluri P, Wilensky RL, Pilla JJ, Gorman JH, Gorman RC, Woo YJ. Preclinical evaluation of the engineered stem cell chemokine stromal cell-derived factor 1α analog in a translational ovine myocardial infarction model. Circ Res 2013; 114:650-9. [PMID: 24366171 DOI: 10.1161/circresaha.114.302884] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
RATIONALE After myocardial infarction, there is an inadequate blood supply to the myocardium, and the surrounding borderzone becomes hypocontractile. OBJECTIVE To develop a clinically translatable therapy, we hypothesized that in a preclinical ovine model of myocardial infarction, the modified endothelial progenitor stem cell chemokine, engineered stromal cell-derived factor 1α analog (ESA), would induce endothelial progenitor stem cell chemotaxis, limit adverse ventricular remodeling, and preserve borderzone contractility. METHODS AND RESULTS Thirty-six adult male Dorset sheep underwent permanent ligation of the left anterior descending coronary artery, inducing an anteroapical infarction, and were randomized to borderzone injection of saline (n=18) or ESA (n=18). Ventricular function, geometry, and regional strain were assessed using cardiac MRI and pressure-volume catheter transduction. Bone marrow was harvested for in vitro analysis, and myocardial biopsies were taken for mRNA, protein, and immunohistochemical analysis. ESA induced greater chemotaxis of endothelial progenitor stem cells compared with saline (P<0.01) and was equivalent to recombinant stromal cell-derived factor 1α (P=0.27). Analysis of mRNA expression and protein levels in ESA-treated animals revealed reduced matrix metalloproteinase 2 in the borderzone (P<0.05), with elevated levels of tissue inhibitor of matrix metalloproteinase 1 and elastin in the infarct (P<0.05), whereas immunohistochemical analysis of borderzone myocardium showed increased capillary and arteriolar density in the ESA group (P<0.01). Animals in the ESA treatment group also had significant reductions in infarct size (P<0.01), increased maximal principle strain in the borderzone (P<0.01), and a steeper slope of the end-systolic pressure-volume relationship (P=0.01). CONCLUSIONS The novel, biomolecularly designed peptide ESA induces chemotaxis of endothelial progenitor stem cells, stimulates neovasculogenesis, limits infarct expansion, and preserves contractility in an ovine model of myocardial infarction.
Collapse
Affiliation(s)
- John W Macarthur
- From the Division of Cardiovascular Surgery, Department of Surgery (J.W.M., J.E.C., J.R.M., A.T., A.S.F., G.H., W.H., A.B.G., P.A., J.H.G., R.C.G.), Gorman Cardiovascular Research Group, Department of Surgery (J.R.M., J.J.P., J.H.G., R.C.G.), Division of Interventional Cardiology, Department of Medicine (R.L.W.), and Department of Radiology (J.J.P.), University of Pennsylvania School of Medicine, Philadelphia; and Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (J.W.M., J.E.C., Y.S., J.B.P., B.B.E., Y.J.W.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shudo Y, Cohen JE, Macarthur JW, Atluri P, Hsiao PF, Yang EC, Fairman AS, Trubelja A, Patel J, Miyagawa S, Sawa Y, Woo YJ. Spatially oriented, temporally sequential smooth muscle cell-endothelial progenitor cell bi-level cell sheet neovascularizes ischemic myocardium. Circulation 2013; 128:S59-68. [PMID: 24030422 DOI: 10.1161/circulationaha.112.000293] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) possess robust therapeutic angiogenic potential, yet may be limited in the capacity to develop into fully mature vasculature. This problem might be exacerbated by the absence of a neovascular foundation, namely pericytes, with simple EPC injection. We hypothesized that coculturing EPCs with smooth muscle cells (SMCs), components of the surrounding vascular wall, in a cell sheet will mimic the native spatial orientation and interaction between EPCs and SMCs to create a supratherapeutic angiogenic construct in a model of ischemic cardiomyopathy. METHODS AND RESULTS Primary EPCs and SMCs were isolated from Wistar rats. Confluent SMCs topped with confluent EPCs were spontaneously detached from the Upcell dish to create an SMC-EPC bi-level cell sheet. A rodent ischemic cardiomyopathy model was created by ligating the left anterior descending coronary artery. Rats were then immediately divided into 3 groups: cell-sheet transplantation (n=14), cell injection (n=12), and no treatment (n=13). Cocultured EPCs and SMCs stimulated an abundant release of multiple cytokines in vitro. Increased capillary density and improved blood perfusion in the borderzone elucidated the significant in vivo angiogenic potential of this technology. Most interestingly, however, cell fate-tracking experiments demonstrated that the cell-sheet EPCs and SMCs directly migrated into the myocardium and differentiated into elements of newly formed functional vasculature. The robust angiogenic effect of this cell sheet translated to enhanced ventricular function as demonstrated by echocardiography. CONCLUSIONS Spatially arranged EPC-SMC bi-level cell-sheet technology facilitated the natural interaction between EPCs and SMCs, thereby creating structurally mature, functional microvasculature in a rodent ischemic cardiomyopathy model, leading to improved myocardial function.
Collapse
Affiliation(s)
- Yasuhiro Shudo
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA (Y.S., J.E.C., J.W.M., P.A., P.F.H., E.C.Y., A.S.F., A.T., J.P., Y.J.W.); and Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan (S.M., Y.S.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Atluri P, Trubelja A, Fairman AS, Hsiao P, MacArthur JW, Cohen JE, Shudo Y, Frederick JR, Woo YJ. Normalization of postinfarct biomechanics using a novel tissue-engineered angiogenic construct. Circulation 2013; 128:S95-104. [PMID: 24030426 DOI: 10.1161/circulationaha.112.000368] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cell-mediated angiogenic therapy for ischemic heart disease has had disappointing results. The lack of clinical translatability may be secondary to cell death and systemic dispersion with cell injection. We propose a novel tissue-engineered therapy, whereby extracellular matrix scaffold seeded with endothelial progenitor cells (EPCs) can overcome these limitations using an environment in which the cells can thrive, enabling an insult-free myocardial cell delivery to normalize myocardial biomechanics. METHODS AND RESULTS EPCs were isolated from the long bones of Wistar rat bone marrow. The cells were cultured for 7 days in media or seeded at a density of 5 × 10(6) cells/cm(2) on a collagen/vitronectin matrix. Seeded EPCs underwent ex vivo modification with stromal cell-derived factor-1α (100 ng/mL) to potentiate angiogenic properties and enhance paracrine qualities before construct formation. Scanning electron microscopy and confocal imaging confirmed EPC-matrix adhesion. In vitro vasculogenic potential was assessed by quantifying EPC cell migration and vascular differentiation. There was a marked increase in vasculogenesis in vitro as measured by angiogenesis assay (8 versus 0 vessels/hpf; P=0.004). The construct was then implanted onto ischemic myocardium in a rat model of acute myocardial infarction. Confocal microscopy demonstrated a significant migration of EPCs from the construct to the myocardium, suggesting a direct angiogenic effect. Myocardial biomechanical properties were uniaxially quantified by elastic modulus at 5% to 20% strain. Myocardial elasticity normalized after implant of our tissue-engineered construct (239 kPa versus normal=193, P=0.1; versus infarct=304 kPa, P=0.01). CONCLUSIONS We demonstrate restoration and normalization of post-myocardial infarction ventricular biomechanics after therapy with an angiogenic tissue-engineered EPC construct.
Collapse
Affiliation(s)
- Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hoh BL, Hosaka K, Downes DP, Nowicki KW, Wilmer EN, Velat GJ, Scott EW. Stromal cell-derived factor-1 promoted angiogenesis and inflammatory cell infiltration in aneurysm walls. J Neurosurg 2013; 120:73-86. [PMID: 24160472 DOI: 10.3171/2013.9.jns122074] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECT A small percentage of cerebral aneurysms rupture, but when they do, the effects are devastating. Current management of unruptured aneurysms consists of surgery, endovascular treatment, or watchful waiting. If the biology of how aneurysms grow and rupture were better known, a novel drug could be developed to prevent unruptured aneurysms from rupturing. Ruptured cerebral aneurysms are characterized by inflammation-mediated wall remodeling. The authors studied the role of stromal cell-derived factor-1 (SDF-1) in inflammation-mediated wall remodeling in cerebral aneurysms. METHODS Human aneurysms, murine carotid artery aneurysms, and murine intracranial aneurysms were studied using immunohistochemistry. Flow cytometry analysis was performed on blood from mice developing carotid or intracranial aneurysms. The effect of SDF-1 on endothelial cells and macrophages was studied by chemotaxis cell migration assay and capillary tube formation assay. Anti-SDF-1 blocking antibody was given to mice and compared with control (vehicle)-administered mice for its effects on the walls of carotid aneurysms and the development of intracranial aneurysms. RESULTS Human aneurysms, murine carotid aneurysms, and murine intracranial aneurysms all expressed SDF-1, and mice with developing carotid or intracranial aneurysms had increased progenitor cells expressing CXCR4, the receptor for SDF-1 (p < 0.01 and p < 0.001, respectively). Human aneurysms and murine carotid aneurysms had endothelial cells, macrophages, and capillaries in the walls of the aneurysms, and the presence of capillaries in the walls of human aneurysms was associated with the presence of macrophages (p = 0.01). Stromal cell-derived factor-1 promoted endothelial cell and macrophage migration (p < 0.01 for each), and promoted capillary tube formation (p < 0.001). When mice were given anti-SDF-1 blocking antibody, there was a significant reduction in endothelial cells (p < 0.05), capillaries (p < 0.05), and cell proliferation (p < 0.05) in the aneurysm wall. Mice given anti-SDF-1 blocking antibody developed significantly fewer intracranial aneurysms (33% vs 89% in mice given control immunoglobulin G, respectively; p < 0.05). CONCLUSIONS These data suggest SDF-1 is associated with angiogenesis and inflammatory cell migration and proliferation in the walls of aneurysms, and may have a role in the development of intracranial aneurysms.
Collapse
|
43
|
du Pré BC, Doevendans PA, van Laake LW. Stem cells for cardiac repair: an introduction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:186-97. [PMID: 23888179 PMCID: PMC3708059 DOI: 10.3969/j.issn.1671-5411.2013.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 02/16/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease is a major cause of morbidity and mortality throughout the world. Most cardiovascular diseases, such as ischemic heart disease and cardiomyopathy, are associated with loss of functional cardiomyocytes. Unfortunately, the heart has a limited regenerative capacity and is not able to replace these cardiomyocytes once lost. In recent years, stem cells have been put forward as a potential source for cardiac regeneration. Pre-clinical studies that use stem cell-derived cardiac cells show promising results. The mechanisms, though, are not well understood, results have been variable, sometimes transient in the long term, and often without a mechanistic explanation. There are still several major hurdles to be taken. Stem cell-derived cardiac cells should resemble original cardiac cell types and be able to integrate in the damaged heart. Integration requires administration of stem cell-derived cardiac cells at the right time using the right mode of delivery. Once delivered, transplanted cells need vascularization, electrophysiological coupling with the injured heart, and prevention of immunological rejection. Finally, stem cell therapy needs to be safe, reproducible, and affordable. In this review, we will give an introduction to the principles of stem cell based cardiac repair.
Collapse
Affiliation(s)
- Bastiaan C du Pré
- Departments of Cardiology and Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, P.O. box 85500, 3508 GA Utrecht, the Netherlands
| | | | | |
Collapse
|
44
|
Diastolic ventricular support with cardiac support devices: an alternative approach to prevent adverse ventricular remodeling. Heart Fail Rev 2013; 18:55-63. [PMID: 22527015 DOI: 10.1007/s10741-012-9312-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Heart failure is a global epidemic with limited therapy. Abnormal left ventricular wall stress in the diseased myocardium results in a biochemical positive feedback loop that results in global ventricular remodeling and further deterioration of myocardial function. Mechanical myocardial restraints such as the Acorn CorCap and Paracor HeartNet ventricular restraints have attempted to minimize diastolic ventricular wall stress and limit adverse ventricular remodeling. Unfortunately, these therapies have not yielded viable clinical therapies for heart failure. Cellular and novel biopolymer-based therapies aimed at stabilizing pathologic myocardium hold promise for translation to clinical therapy in the future.
Collapse
|
45
|
Taghavi S, George JC. Homing of stem cells to ischemic myocardium. Am J Transl Res 2013; 5:404-411. [PMID: 23724164 PMCID: PMC3665914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 05/18/2013] [Indexed: 06/02/2023]
Abstract
Progenitor cells have the capability to home myocardium in response to ischemia. Cell adhesion markers, in particular integrins, play an important role in the trafficking of stem cells to myocardium. In addition, damaged myocardium secretes several chemokines and growth factors that recruit these precursor cells to the heart. Nitric oxide synthase and hormones can also contribute to the trafficking of progenitor cells to myocardium. The recruitment of stem cells to ischemic myocardium is a complex interchange between cell adhesion markers, chemokines, and growth factors and a better understanding of these processes may lead to more efficient use of stem cells for therapeutic benefit.
Collapse
Affiliation(s)
- Sharven Taghavi
- Cardiovascular Research Center, Temple UniversityPhiladelphia, PA, USA
- Hospital Department of Surgery, Temple UniversityPhiladelphia, PA, USA
| | - Jon C George
- Cardiovascular Research Center, Temple UniversityPhiladelphia, PA, USA
- School of Medicine, Temple UniversityPhiladelphia, PA, USA
| |
Collapse
|
46
|
Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 2013; 7:833-50. [PMID: 23164083 DOI: 10.2217/rme.12.82] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue repair and regeneration are thought to involve resident cell proliferation as well as the selective recruitment of circulating stem and progenitor cell populations through complex signaling cascades. Many of these recruited cells originate from the bone marrow, and specific subpopulations of bone marrow cells have been isolated and used to augment adult tissue regeneration in preclinical models. Clinical studies of cell-based therapies have reported mixed results, however, and a variety of approaches to enhance the regenerative capacity of stem cell therapies are being developed based on emerging insights into the mechanisms of progenitor cell biology and recruitment following injury. This article discusses the function and mechanisms of recruitment of important bone marrow-derived stem and progenitor cell populations following injury, as well as the emerging therapeutic applications targeting these cells.
Collapse
Affiliation(s)
- Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Hagey Building GK-201, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
47
|
Lam MT, Wu JC. Biomaterial applications in cardiovascular tissue repair and regeneration. Expert Rev Cardiovasc Ther 2013; 10:1039-49. [PMID: 23030293 DOI: 10.1586/erc.12.99] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cardiovascular disease physically damages the heart, resulting in loss of cardiac function. Medications can help alleviate symptoms, but it is more beneficial to treat the root cause by repairing injured tissues, which gives patients better outcomes. Besides heart transplants, cardiac surgeons use a variety of methods for repairing different areas of the heart such as the ventricular septal wall and valves. A multitude of biomaterials are used in the repair and replacement of impaired heart tissues. These biomaterials fall into two main categories: synthetic and natural. Synthetic materials used in cardiovascular applications include polymers and metals. Natural materials are derived from biological sources such as human donor or harvested animal tissues. A new class of composite materials has emerged to take advantage of the benefits of the strengths and minimize the weaknesses of both synthetic and natural materials. This article reviews the current and prospective applications of biomaterials in cardiovascular therapies.
Collapse
Affiliation(s)
- Mai T Lam
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Hagey Pediatric Regenerative Research Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
48
|
Rehmannia glutinosa extract activates endothelial progenitor cells in a rat model of myocardial infarction through a SDF-1 α/CXCR4 cascade. PLoS One 2013; 8:e54303. [PMID: 23349848 PMCID: PMC3548813 DOI: 10.1371/journal.pone.0054303] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/10/2012] [Indexed: 12/24/2022] Open
Abstract
Objectives Endothelial progenitor cells (EPCs) can be used to repair tissues after myocardial infarction (MI) but EPC activators have adverse reactions. Rehmannia glutinosa is a herb used in traditional Chinese medicine, which can promote bone-marrow proliferation and protect the ischemic myocardium. We investigated the effects of Rehmannia glutinosa extract (RGE) on EPCs in a rat model of MI. Methods A total of 120 male Wistar rats were randomized to 2 groups (n = 60 each) for treatment: high-dose RGE (1.5 g·kg−1·day−1 orally) for 8 weeks, then left anterior descending coronary artery ligation, mock surgery or no treatment, then RGE orally for 4 weeks; or normal saline (NS) as the above protocol. The infarct region of the left ventricle was assessed by serial sectioning and morphology. EPCs were evaluated by number and function. Protein and mRNA levels of CD133, vascular endothelial growth factor receptor 2 (VEGFR2), chemokine C-X-C motif receptor 4 (CXCR4), stromal cell–derived factor-1α (SDF-1α) were measured by immunohistochemistry, Western blot and quantitative PCR analysis. Results RGE significantly improved left ventricular function, decreased the ischemic area and the apoptotic index in the infarct myocardium, also decreased the concentration of serum cardiac troponin T and brain natriuretic peptide at the chronic stage after MI (from week 2 to week 4). RGE increased EPC number, proliferation, migration and tube-formation capacity. It was able to up-regulate the expression of angiogenesis-associated ligand/receptor, including CD133, VEGFR2 and SDF-1α/CXCR4. In vitro, the effect of RGE on SDF-1α/CXCR4 cascade was reversed by the CXCR4 specific antagonist AMD3100. Conclusion RGE may enhance the mobilization, migration and therapeutic angiogenesis of EPCs after MI by activating the SDF-1α/CXCR4 cascade.
Collapse
|
49
|
Monaghan M, Greiser U, Wall JG, O’Brien T, Pandit A. Interference: an alteRNAtive therapy following acute myocardial infarction. Trends Pharmacol Sci 2012; 33:635-45. [DOI: 10.1016/j.tips.2012.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
|
50
|
Association of stromal-derived factor-1 alpha and endogenous sex hormones in men aged over 50 years with stable coronary artery disease. Adv Med Sci 2012. [PMID: 23192056 DOI: 10.2478/v10039-012-0051-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE Many studies indicate an inverse relationship between stromal-derived factor-1 alpha (SDF-1 alpha), a chemokine, and coronary risk factors. Moreover, SDF-1 alpha is crucial in neoangiogenesis and in the mobilization and homing of endothelial progenitor cells to the ischemic coronary vessels. Numerous studies indicate that circulating sex hormones are associated with atherogenesis during male aging. The aim of this study was therefore to determine whether there exists a relationship between SDF-1 alpha and endogenous sex hormones in aging men with stable coronary artery disease (CAD). MATERIAL AND METHODS Plasma concentrations of SDF-1 alpha, testosterone (T), estradiol (E2), and sex hormone binding globulin (SHBG) were measured and the E2/T ratio was calculated in a cross-sectional study of 82 men over 50 years of age with stable CAD. RESULTS SDF-1 alpha was positively and significantly correlated with T (r = 0.233; p = 0.036) and with SHBG (r = 0.312; p = 0.004). There was a significant inverse correlation between SDF-1 alpha and the E2/T ratio (r = -0.463; p < 0.001). After adjustment for age, body mass index and smoking status, SHBG and E2/T ratio were the only factors associated with SDF-1 alpha. CONCLUSIONS T and SHBG (directly) and the E2/T ratio (inversely) may be involved in the etiopathogenesis of CAD through their relationships to SDF-1 alpha.
Collapse
|