1
|
Yao J, Ma F, Zhang L, Zhu C, Jumabay M, Yao Z, Wang L, Cai X, Zhang D, Qiao X, Shivkumar K, Pellegrini M, Yao Y, Wu X, Boström KI. Single-Cell RNA-Seq Identifies Dynamic Cardiac Transition Program from Adipose Derived Cells Induced by Leukemia Inhibitory Factor. Stem Cells 2022; 40:932-948. [PMID: 35896368 PMCID: PMC9585902 DOI: 10.1093/stmcls/sxac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022]
Abstract
Adipose-derived cells (ADCs) from white adipose tissue (WAT) are promising stem cell candidates because of their large regenerative reserves and the potential for cardiac regeneration. However, given the heterogeneity of ADC and its unsolved mechanisms of cardiac acquisition, ADC-cardiac transition efficiency remains low. In this study, we explored the heterogeneity of ADCs and the cellular kinetics of 39,432 single-cell transcriptomes along the leukemia inhibitory factor (LIF) induced ADC-cardiac transition. We identified distinct ADC subpopulations that reacted differentially to LIF when entering the cardiomyogenic program, further demonstrating that ADC-myogenesis is time-dependent and initiates from transient changes in nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. At later stages, pseudotime analysis of ADCs navigated a trajectory with two branches corresponding to activated myofibroblast or cardiomyocyte-like cells. Our findings offer a high-resolution dissection of ADC heterogeneity and cell fate during ADC-cardiac transition, thus providing new insights into potential cardiac stem cells.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Feiyang Ma
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Chongqing International Institute for Immunology, Chongqing 401338, China
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Ching Zhu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Medet Jumabay
- Division of Allergy, Immunology Center for Immunity, Infection, and Inflammation Pediatrics, Dept of Medicine, University of California, San Diego, San Diego, CA
| | - Zehao Yao
- Peking Union Medical College, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Lumin Wang
- Institute of Precision Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Daoqin Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | | | - Matteo Pellegrini
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Dept of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
2
|
Narita S, Unno K, Kato K, Okuno Y, Sato Y, Tsumura Y, Fujikawa Y, Shimizu Y, Hayashida R, Kondo K, Shibata R, Murohara T. Direct reprogramming of adult adipose-derived regenerative cells toward cardiomyocytes using six transcriptional factors. iScience 2022; 25:104651. [PMID: 35811849 PMCID: PMC9263527 DOI: 10.1016/j.isci.2022.104651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 04/30/2022] [Accepted: 06/16/2022] [Indexed: 10/29/2022] Open
|
3
|
Polymers and Nanoparticles for Statin Delivery: Current Use and Future Perspectives in Cardiovascular Disease. Polymers (Basel) 2021; 13:polym13050711. [PMID: 33652927 PMCID: PMC7956757 DOI: 10.3390/polym13050711] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis-related coronary artery disease (CAD) is one of the leading sources of mortality and morbidity in the world. Primary and secondary prevention appear crucial to reduce CAD-related complications. In this scenario, statin treatment was shown to be clinically effective in the reduction of adverse events, but systemic administration provides suboptimal results. As an attempt to improve bioavailability and effectiveness, polymers and nanoparticles for statin delivery were recently investigated. Polymers and nanoparticles can help statin delivery and their effects by increasing oral bioavailability or enhancing target-specific interaction, leading to reduced vascular endothelial dysfunction, reduced intimal hyperplasia, reduced ischemia-reperfusion injury, increased cardiac regeneration, positive remodeling in the extracellular matrix, reduced neointimal growth and increased re-endothelization. Moreover, some innovative aspects described in other cardiovascular fields could be translated into the CAD scenario. Recent preclinical studies are underlining the effect of statins in the stimulation and differentiation of endogenous cardiac stem cells, as well as in targeting of local adverse conditions implicated in atherosclerosis, and statin delivery through poly-lactic-co-glycolic acid (PLGA) appears the most promising aspect of current research to enhance drug activity. The present review intends to summarize the current evidence about polymers and nanoparticles for statin delivery in the field of cardiovascular disease, trying to shed light on this topic and identify new avenues for future studies.
Collapse
|
4
|
Fujiwara O, Prasai A, Perez-Bello D, El Ayadi A, Petrov IY, Esenaliev RO, Petrov Y, Herndon DN, Finnerty CC, Prough DS, Enkhbaatar P. Adipose-derived stem cells improve grafted burn wound healing by promoting wound bed blood flow. BURNS & TRAUMA 2020; 8:tkaa009. [PMID: 32346539 PMCID: PMC7175768 DOI: 10.1093/burnst/tkaa009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/22/2020] [Accepted: 01/30/2000] [Indexed: 01/08/2023]
Abstract
BACKGROUND Researchers have explored the use of adipose-derived stem cells (ASCs) as a cell-based therapy to cover wounds in burn patients; however, underlying mechanistic aspects are not completely understood. We hypothesized that ASCs would improve post-burn wound healing after eschar excision and grafting by increasing wound blood flow via induction of angiogenesis-related pathways. METHODS To test the hypothesis, we used an ovine burn model. A 5 cm2 full thickness burn wound was induced on each side of the dorsum. After 24 hours, the burned skin was excised and a 2 cm2 patch of autologous donor skin was grafted. The wound sites were randomly allocated to either topical application of 7 million allogeneic ASCs or placebo treatment (phosphate-buffered saline [PBS]). Effects of ASCs culture media was also compared to those of PBS. Wound healing was assessed at one and two weeks following the application of ASCs. Allogeneic ASCs were isolated, cultured and characterized from non-injured healthy sheep. The identity of the ASCs was confirmed by flow cytometry analysis, differentiation into multiple lineages and gene expression via real-time polymerase chain reaction. Wound blood flow, epithelialization, graft size and take and the expression of vascular endothelial growth factor (VEGF) were determined via enzyme-linked immunosorbent assay and Western blot. RESULTS Treatment with ASCs accelerated the patch graft growth compared to the control (p < 0.05). Topical application of ASCs significantly increased wound blood flow (p < 0.05). Expression of VEGF was significantly higher in the wounds treated with ASCs compared to control (p < 0.05). CONCLUSIONS ASCs accelerated grafted skin growth possibly by increasing the blood flow via angiogenesis induced by a VEGF-dependent pathway.
Collapse
Affiliation(s)
- Osamu Fujiwara
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Anesh Prasai
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| | - Dannelys Perez-Bello
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Amina El Ayadi
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
- Sealy Center for Molecular Medicine, and the Institute for Translational Sciences, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Irene Y Petrov
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
| | - Rinat O Esenaliev
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Yuriy Petrov
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
| | - David N Herndon
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| | - Celeste C Finnerty
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
- Sealy Center for Molecular Medicine, and the Institute for Translational Sciences, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| |
Collapse
|
5
|
Atkinson SP. Previews. Stem Cells Transl Med 2019. [PMCID: PMC6766597 DOI: 10.1002/sctm.19-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
6
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
7
|
Yokoyama R, Ii M, Masuda M, Tabata Y, Hoshiga M, Ishizaka N, Asahi M. Cardiac Regeneration by Statin-Polymer Nanoparticle-Loaded Adipose-Derived Stem Cell Therapy in Myocardial Infarction. Stem Cells Transl Med 2019; 8:1055-1067. [PMID: 31157513 PMCID: PMC6766602 DOI: 10.1002/sctm.18-0244] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 04/20/2019] [Indexed: 12/15/2022] Open
Abstract
Clinical trials with autologous adipose‐derived stem cell (AdSC) therapy for ischemic heart diseases (IHDs) are ongoing. However, little is known about combinational therapeutic effect of AdSCs and statin poly(lactic‐co‐glycolic) acid (PLGA) nanoparticles on the ischemic myocardium. We investigated the hypothesis that statins, which have pleiotropic effects, augment the therapeutic potential of AdSCs and that AdSCs also act as drug delivery tools. Simvastatin‐conjugated nanoparticles (SimNPs) significantly promoted migration activity without changing proliferation activity and upregulated growth factor gene expression in vitro. A small number of intravenously administered SimNP‐loaded AdSCs (10,000 cells per mouse) improved cardiac function following myocardial infarction, inducing endogenous cardiac regeneration in the infarcted myocardium. The de novo regenerated myocardium was thought to be derived from epicardial cells, which were positive for Wilms' tumor protein 1 expression. These findings were attributed to the sustained, local simvastatin release from the recruited SimNP‐loaded AdSCs in the infarcted myocardium rather than to the direct contribution of recruited AdSCs to tissue regeneration. SimNP‐loaded AdSCs may lead to a novel somatic stem cell therapy for IHDs. stem cells translational medicine2019;8:1055–1067
Collapse
Affiliation(s)
- Ryo Yokoyama
- Department of Cardiology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Ii
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, Osaka, Japan
| | - Misaki Masuda
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, Osaka, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Nobukazu Ishizaka
- Department of Cardiology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| |
Collapse
|
8
|
Long-Term Biobanking of Intact Tissue from Lipoaspirate. J Clin Med 2019; 8:jcm8030327. [PMID: 30857129 PMCID: PMC6463172 DOI: 10.3390/jcm8030327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/14/2019] [Accepted: 02/26/2019] [Indexed: 01/09/2023] Open
Abstract
Autologous fat grafting has now been extensively and successfully performed for more than two decades. Although most adipose grafts and adipose-derived MSC therapies are done with fresh tissue, cryopreservation of tissue allows for much greater flexibility of use. Over the course of five years, 194 cryopreserved adipose samples were thawed and then returned to the collecting physician for subsequent autologous applications. Samples were stored with a mean cryogenic storage time of 9.5 months, with some samples being stored as long as 44 months. The volumes of tissue stored varied from 12 cc to as large as 960 cc. Upon thawing, the volume of recovered whole adipose tissue averaged 67% of the original amount stored for all samples, while the samples that were stored for longer than one year averaged 71%. Recovery was not found to be a function of length of time in cryopreservation. No significant relationship was found between tissue recovery and patient age. While an average recovery of 67% of volume frozen indicates that the use of banked and thawed tissue requires a larger amount of sample to be taken from the patient initially, an experienced clinician easily accomplishes this requirement. As cryopreservation of adipose tissue becomes more commonplace, physicians will find it helpful to know the amount and quality of tissue that will be available after thawing procedures.
Collapse
|
9
|
Chen CF, Liao HT. Platelet-rich plasma enhances adipose-derived stem cell-mediated angiogenesis in a mouse ischemic hindlimb model. World J Stem Cells 2018; 10:212-227. [PMID: 30613314 PMCID: PMC6306556 DOI: 10.4252/wjsc.v10.i12.212] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/18/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the angiogenic effect of platelet-rich plasma (PRP)-preconditioned adipose-derived stem cells (ADSCs) both in vitro and in a mouse ischemic hindlimb model.
METHODS ADSCs were divided based on culture medium: 2.5% PRP, 5% PRP, 7.5% PRP, and 10% PRP. Cell proliferation rate was analyzed using the MTS assay. The gene expression of CD31, vascular endothelial growth factor, hypoxia-inducible factors, and endothelial cell nitric oxide synthase was analyzed using reverse transcription polymerase chain reaction. Cell markers and structural changes were assessed through immunofluorescence staining and the tube formation assay. Subsequently, we studied the in vivo angiogenic capabilities of ADSCs by a mouse ischemic hindlimb model.
RESULTS The proliferation rate of ADSCs was higher in the 2.5%, 5%, and 7.5% PRP groups. The expression of hypoxia-inducible factor, CD31, vascular endothelial growth factor, and endothelial cell nitric oxide synthase in the 5% and 7.5% PRP groups increased. The 5%, 7.5%, and 10% PRP groups showed higher abilities to promote both CD31 and vascular endothelial growth factor production and tubular structure formation in ADSCs. According to laser Doppler perfusion scan, the perfusion ratios of ischemic limb to normal limb were significantly higher in 5% PRP, 7.5% PRP, and human umbilical vein endothelial cells groups compared with the negative control and fetal bovine serum (FBS) groups (0.88 ± 0.08, 0.85 ± 0.07 and 0.81 ± 0.06 for 5%, 7.5% PRP and human umbilical vein endothelial cells compared with 0.42 ± 0.17 and 0.54 ± 0.14 for the negative control and FBS, P < 0.01).
CONCLUSION PRP-preconditioned ADSCs presented endothelial cell characteristics in vitro and significantly improved neovascularization in ischemic hindlimbs. The optimal angiogenic effect occurred in 5% PRP- and 7.5% PRP-preconditioned ADSCs.
Collapse
Affiliation(s)
- Chia-Fang Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Han-Tsung Liao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Craniofacial Research Center, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
10
|
Luo L, Zheng W, Lian G, Chen H, Li L, Xu C, Xie L. Combination treatment of adipose-derived stem cells and adiponectin attenuates pulmonary arterial hypertension in rats by inhibiting pulmonary arterial smooth muscle cell proliferation and regulating the AMPK/BMP/Smad pathway. Int J Mol Med 2017; 41:51-60. [PMID: 29115380 PMCID: PMC5746303 DOI: 10.3892/ijmm.2017.3226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to assess the effects of therapy with adiponectin (APN) gene-modified adipose-derived stem cells (ADSCs) on pulmonary arterial hypertension (PAH) in rats and the underlying cellular and molecular mechanisms. ADSCs were successfully isolated from the rats and characterized. ADSCs were effectively infected with the green fluorescent protein (GFP)-empty (ADSCs-V) or the APN-GFP (ADSCs-APN) lentivirus and the APN expression was evaluated by ELISA. Sprague-Dawley rats were administered monocrotaline (MCT) to develop PAH. The rats were treated with MCT, ADSCs, ADSCs-V and ADSCs-APN. Then ADSCs-APN in the lung were investigated by confocal laser scanning microscopy and western blot analysis. Engrafted ADSCs in the lung were located around the vessels. Mean pulmonary arterial pressure (mPAP) and the right ventricular hypertrophy index (RVHI) in the ADSCs-APN-treated mice were significantly decreased as compared with the ADSCs and ADSCs-V treatments. Pulmonary vascular remodeling was assessed. Right ventricular (RV) function was evaluated by echocardiography. We found that pulmonary vascular remodeling and the parameters of RV function were extensively improved after ADSCs-APN treatment when compared with ADSCs and ADSCs-V treatment. Pulmonary artery smooth muscle cells (PASMCs) were isolated from the PAH rats. The antiproliferative effect of APN on PASMCs was assayed by Cell Counting Kit-8. The influence of APN and specific inhibitors on the levels of bone morphogenetic protein (BMP), adenosine monophosphate activated protein kinase (AMPK), and small mothers against decapentaplegia (Smad) pathways was detected by western blot analysis. We found that APN suppressed the proliferation of PASMCs isolated from the PAH rats by regulating the AMPK/BMP/Smad pathway. This effect was weakened by addition of the AMPK inhibitor (compound C) and BMP2 inhibitor (noggin). Therefore, combination treatment with ADSCs and APN effectively attenuated PAH in rats by inhibiting PASMC proliferation and regulating the AMPK/BMP/Smad pathway.
Collapse
Affiliation(s)
- Li Luo
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Wuhong Zheng
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Guili Lian
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Huaning Chen
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ling Li
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Changsheng Xu
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Liangdi Xie
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
11
|
Lukovic D, Zlabinger K, Gugerell A, Spannbauer A, Pavo N, Mandic L, Weidenauer DT, Kastl S, Kaun C, Posa A, Sabdyusheva Litschauer I, Winkler J, Gyöngyösi M. Inhibition of CD34+ cell migration by matrix metalloproteinase-2 during acute myocardial ischemia, counteracted by ischemic preconditioning. F1000Res 2016; 5:2739. [PMID: 28299177 PMCID: PMC5321121 DOI: 10.12688/f1000research.9957.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
Background. Mobilization of bone marrow-origin CD34+ cells was investigated 3 days (3d) after acute myocardial infarction (AMI) with/without ischemic preconditioning (IP) in relation to stromal-derived factor-1 (SDF-1α)/ chemokine receptor type 4 (CXCR4) axis, to search for possible mechanisms behind insufficient cardiac repair in the first days post-AMI.
Methods. Closed-chest reperfused AMI was performed by percutaneous balloon occlusion of the mid-left anterior descending (LAD) coronary artery for 90min, followed by reperfusion in pigs. Animals were randomized to receive either IP initiated by 3x5min cycles of re-occlusion/re-flow prior to AMI (n=6) or control AMI (n=12). Blood samples were collected at baseline, 3d post-AMI, and at 1-month follow-up to analyse chemokines and mobilized CD34+ cells. To investigate the effect of acute hypoxia, SDF-1α and matrix metalloproteinase (MMP)-2
in vitro were assessed, and a migration assay of CD34+ cells toward cardiomyocytes was performed.
Results. Reperfused AMI induced significant mobilisation of CD34+ cells (baseline: 260±75 vs. 3d: 668±180; P<0.001) and secretion of MMP-2 (baseline: 291.83±53.40 vs. 3d: 369.64±72.89; P=0.011) into plasma, without affecting the SDF-1α concentration. IP led to the inhibition of MMP-2 (IP: 165.67±47.99 vs. AMI: 369.64±72.89; P=0.004) 3d post-AMI, accompanied by increased release of SDF-1α (baseline: 23.80±12.36 vs. 3d: 45.29±11.31; P=0.05) and CXCR4 (baseline: 0.59±0.16 vs. 3d: 2.06±1.42; P=0.034), with a parallel higher level of mobilisation of CD34+ cells (IP: 881±126 vs. AMI: 668±180; P=0.026), compared to non-conditioned AMI.
In vitro, CD34+ cell migration toward cardiomyocytes was enhanced by SDF-1α, which was completely abolished by 90min hypoxia and co-incubation with MMP-2.
Conclusions. Non-conditioned AMI induces MMP-2 release, hampering the ischemia-induced increase in SDF-1α and CXCR4 by cleaving the SDF-1α/CXCR4 axis, with diminished mobilization of the angiogenic CD34+ cells. IP might influence CD34+ cell mobilization via inhibition of MMP-2.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Ljubica Mandic
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Stefan Kastl
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christoph Kaun
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Aniko Posa
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Nagata H, Ii M, Kohbayashi E, Hoshiga M, Hanafusa T, Asahi M. Cardiac Adipose-Derived Stem Cells Exhibit High Differentiation Potential to Cardiovascular Cells in C57BL/6 Mice. Stem Cells Transl Med 2015; 5:141-51. [PMID: 26683873 DOI: 10.5966/sctm.2015-0083] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022] Open
Abstract
Adipose-derived stem cells (AdSCs) have recently been shown to differentiate into cardiovascular lineage cells. However, little is known about the fat tissue origin-dependent differences in AdSC function and differentiation potential. AdSC-rich cells were isolated from subcutaneous, visceral, cardiac (CA), and subscapular adipose tissue from mice and their characteristics analyzed. After four different AdSC types were cultured with specific differentiation medium, immunocytochemical analysis was performed for the assessment of differentiation into cardiovascular cells. We then examined the in vitro differentiation capacity and therapeutic potential of AdSCs in ischemic myocardium using a mouse myocardial infarction model. The cell density and proliferation activity of CA-derived AdSCs were significantly increased compared with the other adipose tissue-derived AdSCs. Immunocytochemistry showed that CA-derived AdSCs had the highest appearance rates of markers for endothelial cells, vascular smooth muscle cells, and cardiomyocytes among the AdSCs. Systemic transfusion of CA-derived AdSCs exhibited the highest cardiac functional recovery after myocardial infarction and the high frequency of the recruitment to ischemic myocardium. Moreover, long-term follow-up of the recruited CA-derived AdSCs frequently expressed cardiovascular cell markers compared with the other adipose tissue-derived AdSCs. Cardiac adipose tissue could be an ideal source for isolation of therapeutically effective AdSCs for cardiac regeneration in ischemic heart diseases. Significance: The present study found that cardiac adipose-derived stem cells have a high potential to differentiate into cardiovascular lineage cells (i.e., cardiomyocytes, endothelial cells, and vascular smooth muscle cells) compared with stem cells derived from other adipose tissue such as subcutaneous, visceral, and subscapular adipose tissue. Notably, only a small number of supracardiac adipose-derived stem cells that were systemically transplanted sufficiently improved cardiac functional recovery after myocardial infarction, differentiating into cardiovascular cells in the ischemic myocardium. These findings suggest a new autologous stem cell therapy for patients with myocardial ischemia, especially those with secondary myocardial ischemia after cardiovascular open chest surgery.
Collapse
Affiliation(s)
- Hiroki Nagata
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Ii
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Eiko Kohbayashi
- Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Internal Medicine (III), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Toshiaki Hanafusa
- Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| |
Collapse
|
13
|
Reply: The Effect of Adipose-Derived Stem Cell Supernatant on Reperfusion Injury. Plast Reconstr Surg 2015; 136:848e-849e. [PMID: 26322808 DOI: 10.1097/prs.0000000000001753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Adipose-derived Mesenchymal Stem Cells and Their Reparative Potential in Ischemic Heart Disease. ACTA ACUST UNITED AC 2015; 68:599-611. [DOI: 10.1016/j.rec.2015.02.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
|
15
|
Badimon L, Oñate B, Vilahur G. Células madre mesenquimales derivadas de tejido adiposo y su potencial reparador en la enfermedad isquémica coronaria. Rev Esp Cardiol 2015. [DOI: 10.1016/j.recesp.2015.02.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Anderson EM, Mooney DJ. The Combination of Vascular Endothelial Growth Factor and Stromal Cell-Derived Factor Induces Superior Angiogenic Sprouting by Outgrowth Endothelial Cells. J Vasc Res 2015; 52:62-9. [PMID: 26045306 DOI: 10.1159/000382129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 03/30/2015] [Indexed: 11/19/2022] Open
Abstract
Endothelial progenitor cells are being broadly explored for the treatment of ischemic cardiovascular diseases, but their response to molecules commonly used to promote the growth of new blood vessels has not been fully characterized. In this study, angiogenic sprout formation in a 3-dimensional, in vitro model by one type of endothelial progenitor, outgrowth endothelial cells (OECs), was characterized in response to exposure to stromal cell-derived factor (SDF) and vascular endothelial growth factor (VEGF) and then compared to mature endothelial cells. Exposure to SDF alone did not increase angiogenic sprouting in comparison to control media, while a combination of VEGF and SDF demonstrated greater potency than VEGF alone for all cell types. Together, VEGF and SDF reduced the sprout initiation time and maintained sprouting levels over time. In direct competition with mature endothelial cells, OECs preferentially localized to the tip cell position, suggesting an enhanced sprouting potential. Overall, these results reveal the impact of the combination of VEGF and SDF on endothelial cell sprouting, and support the enhanced potential of OECs, as opposed to mature endothelial cells, for treating ischemic diseases.
Collapse
Affiliation(s)
- Erin M Anderson
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Mass., USA
| | | |
Collapse
|
17
|
Minteer DM, Marra KG, Rubin JP. Adipose stem cells: biology, safety, regulation, and regenerative potential. Clin Plast Surg 2015; 42:169-79. [PMID: 25827561 DOI: 10.1016/j.cps.2014.12.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article discusses adipose-derived stem cell (ASC) biology, describes the current knowledge in the literature for the safety and regulation of ASCs, and provides a brief overview of the regenerative potential of ASCs. It is not an exhaustive listing of all available clinical studies or every study applying ASCs in tissue engineering and regenerative medicine, but is an objective commentary of these topics.
Collapse
Affiliation(s)
- Danielle M Minteer
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA.
| |
Collapse
|
18
|
Therapeutic application of adipose derived stem cells in acute myocardial infarction: lessons from animal models. Stem Cell Rev Rep 2014; 10:389-98. [PMID: 24577790 DOI: 10.1007/s12015-014-9502-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The majority of patients survive an acute myocardial infarction (AMI). Their outcome is negatively influenced by post-AMI events, such as loss of viable cardiomyocytes due to a post-AMI inflammatory response, eventually resulting in heart failure and/or death. Recent pre-clinical animal studies indicate that mesenchymal stem cells derived from adipose tissue (ASC) are new promising candidates that may facilitate cardiovascular regeneration in the infarcted myocardium. In this review we have compared all animal studies in which ASC were used as a therapy post-AMI and have focused on aspects that might be important for future successful clinical application of ASC.
Collapse
|
19
|
Nakamura T, Torimura T, Iwamoto H, Kurogi J, Inoue H, Hori Y, Sumie S, Fukushima N, Sakata M, Koga H, Abe M, Ikezono Y, Hashimoto O, Ueno T, Oho K, Okamura T, Okuda S, Kawamoto A, Ii M, Asahara T, Sata M. CD34(+) cell therapy is safe and effective in slowing the decline of hepatic reserve function in patients with decompensated liver cirrhosis. J Gastroenterol Hepatol 2014; 29:1830-8. [PMID: 24731186 DOI: 10.1111/jgh.12622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 μg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.
Collapse
Affiliation(s)
- Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
INTRODUCTION Adipose-derived stem cells (ASCs) have shown potential for cell-based therapy in the field of plastic surgery. However, the fate of ASCs after transplantation and the mechanism(s) of their biologic capabilities remain unclear. METHODS We isolated and cultured ASCs from transgenic mice that express both luciferase and green fluorescent protein and injected the cells into the inguinal fat pads of wild-type mice. We tested 4 experimental groups, namely, ischemic fat pads with/without ASCs and control fat pads with/without ASCs. RESULTS Transplanted ASCs were tracked with bioluminescence imaging. The luminescence gradually decreased over 28 days, indicating cell death after transplantation. More ASCs were retained in ischemic fat pads on day 7 compared to control fat pads. On day 14, adipose tissue vascular density was higher in the ASC transplantation groups compared to those without ASCs. On day 28, there was decreased atrophy of adipose tissue in ASC-treated ischemic fat pads. Transplanted ASCs were detected as nonproliferating green fluorescent protein-positive cells, whereas native endothelial cells adjacent to the transplanted ASCs were proliferative. Protein analysis demonstrated higher expression of hepatocyte growth factor and vascular endothelial growth factor in the ASC transplantation groups, suggesting a paracrine mechanism, which was confirmed by in vitro experiments with conditioned media from ASCs. CONCLUSIONS Transplanted ASCs are preferentially retained in ischemic adipose tissue, although most of the cells eventually undergo cell death. They exert an angiogenic effect on adipose tissue mainly through a paracrine mechanism. Increased understanding of these effects will help develop ASCs as a tool for cell-based therapy.
Collapse
|
21
|
Multilayered adipose-derived regenerative cell sheets created by a novel magnetite tissue engineering method for myocardial infarction. Int J Cardiol 2014; 175:545-53. [DOI: 10.1016/j.ijcard.2014.06.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 04/26/2014] [Accepted: 06/24/2014] [Indexed: 02/08/2023]
|
22
|
Abstract
Adipose-derived stem cells (ASCs) are considered a great alternative source of mesenchymal stem cells (MSCs). Unlike bone marrow stem cells (BMSCs), ASCs can be retrieved in high numbers from lipoaspirate, a by-product of liposuction procedures. Given that ASCs represent an easily accessible and abundant source of multipotent cells, ASCs have garnered attention and curiosity from both scientific and clinical communities for their potential in clinical applications. Furthermore, their unique immunobiology and secretome are attractive therapeutic properties. A decade since the discovery of a stem cell reservoir residing within adipose tissue, ASC-based clinical trials have grown over the years around the world along with assessments made on their safety and efficacy. With the progress of ASCs into clinical applications, the aim towards producing clinical-grade ASCs becomes increasingly important. Several countries have recognised the growing industry of cell therapies and have developed regulatory frameworks to assure their safety. With more research efforts made to understand their effects in both scientific and clinical settings, ASCs hold great promise as a future therapeutic strategy in treating a wide variety of diseases. Therefore, this review seeks to highlight the clinical applicability of ASCs as well as their progress in clinical trials across various medical disciplines.
Collapse
|
23
|
Dai W, Kay GL, Kloner RA. The Therapeutic Effect of Cell Transplantation Versus Noncellular Biomaterial Implantation on Cardiac Structure and Function Following Myocardial Infarction. J Cardiovasc Pharmacol Ther 2014; 19:350-357. [DOI: 10.1177/1074248413517746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although numerous studies demonstrated that localized delivery of either cells or biomaterials improved postinfarction cardiac function, the underlying mechanisms for this effect remain unclear. We performed a comparison of the effects of fetal, neonatal, and human embryonic stem cell-derived cardiac cell as well as mesenchymal stem cell transplantation versus biomaterial (collagen/extracellular matrix) implantation therapy in rat myocardial infarction model in our laboratory, specifically comparing their effects on infarct wall thickness, neovascularization, infarct wall motion, and left ventricular ejection fraction (LVEF). Both cell and biomaterial treatment had similar beneficial effects on cardiac structure (increasing infarct wall thickness and preventing infarct expansion) and function (preventing paradoxical LV systolic bulging and improving LVEF). In this review, we also discussed the underlying mechanisms of cell and biomaterial therapies, their advantages and disadvantages, and future research directions in the field of regenerative cardiology.
Collapse
Affiliation(s)
- Wangde Dai
- The Heart Institute, Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gregory L. Kay
- The Heart Institute, Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert A. Kloner
- The Heart Institute, Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
24
|
Li Q, Zhang A, Tao C, Li X, Jin P. The role of SDF-1-CXCR4/CXCR7 axis in biological behaviors of adipose tissue-derived mesenchymal stem cells in vitro. Biochem Biophys Res Commun 2013; 441:675-80. [PMID: 24184476 DOI: 10.1016/j.bbrc.2013.10.071] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/15/2013] [Indexed: 01/07/2023]
Abstract
Numerous studies have reported that CXCR4 and CXCR7 play an essential, but differential role in stromal cell-derived factor-1 (SDF-1)-inducing cell chemotaxis, viability and paracrine actions of BMSCs. Adipose tissue-derived mesenchymal stem cells (ADSCs) have been suggested to be potential seed cells for clinical application instead of bone marrow derived stroma cell (BMSCs). However, the function of SDF-1/CXCR4 and SDF-1/CXCR7 in ADSCs is not well understood. This study was designed to analyze the effect of SDF-1/CXCR4 and SDF-1/CXCR7 axis on ADSCs biological behaviors in vitro. Using Flow cytometry and Western blot methods, we found for the first time that CXCR4/CXCR7 expression was increased after treatment with SDF-1 in ADSCs. SDF-1 promoted ADSCs paracrine, proliferation and migration abilities. CXCR4 or CXCR7 antibody suppressed ADSCs paracrine action induced by SDF-1. The migration of ADSCs can be abolished by CXCR4 antibody, while the proliferation of ADSCs was only downregulated by CXCR7 antibody. Our study indicated that the angiogenesis of ADSCs is, at least partly, mediated by SDF-1/CXCR4 and SDF-1/CXCR7 axis. However, only binding of SDF-1/CXCR7 was required for proliferation of ADSCs, and CXCR7 was required for migration of ADSCs induced by SDF-1. Our studies provide evidence that the activation of either axis may be helpful to improve the effectiveness of ADSCs-based stem cell therapy.
Collapse
Affiliation(s)
- Qiang Li
- Plastic Surgery Department, Xuzhou Medical College Affiliated Hospital, Jiangsu, China
| | | | | | | | | |
Collapse
|
25
|
Human vasculogenic cells form functional blood vessels and mitigate adverse remodeling after ischemia reperfusion injury in rats. Angiogenesis 2013; 16:773-84. [PMID: 23666122 DOI: 10.1007/s10456-013-9354-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 05/03/2013] [Indexed: 01/30/2023]
Abstract
Cell-based therapies to restore heart function after infarction have been tested in pre-clinical models and clinical trials with mixed results, and will likely require both contractile cells and a vascular network to support them. We and others have shown that human endothelial colony forming cells (ECFC) combined with mesenchymal progenitor cells (MPC) can be used to "bio-engineer" functional human blood vessels. Here we investigated whether ECFC + MPC form functional vessels in ischemic myocardium and whether this affects cardiac function or remodeling. Myocardial ischemia/reperfusion injury (IRI) was induced in 12-week-old immunodeficient rats by ligation of the left anterior descending coronary artery. After 40 min, myocardium was reperfused and ECFC + MPC (2 × 10(6) cells, 2:3 ratio) or PBS was injected. Luciferase assays after injection of luciferase-labeled ECFC + MPC showed that 1,500 ECFC were present at day 14. Human ECFC-lined perfused vessels were directly visualized by femoral vein injection of a fluorescently-tagged human-specific lectin in hearts injected with ECFC + MPC but not PBS alone. While infarct size at day 1 was no different, LV dimensions and heart weight to tibia length ratios were lower in cell-treated hearts compared with PBS at 4 months, suggesting post-infarction remodeling was ameliorated by local cell injection. Fractional shortening, LV wall motion score, and fibrotic area were not different between groups at 4 months. However, pressure-volume loops demonstrated improved cardiac function and reduced volumes in cell-treated animals. These data suggest that myocardial delivery of ECFC + MPC at reperfusion may provide a therapeutic strategy to mitigate LV remodeling and cardiac dysfunction after IRI.
Collapse
|
26
|
Analysis for apoptosis and necrosis on adipocytes, stromal vascular fraction, and adipose-derived stem cells in human lipoaspirates after liposuction. Plast Reconstr Surg 2013; 131:77e-85e. [PMID: 23271558 DOI: 10.1097/prs.0b013e3182729ff7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Adipose-derived stem cells have become the most studied adult stem cells. The authors examined the apoptosis and necrosis rates for adipocyte, stromal vascular fraction, and adipose-derived stem cells in fresh human lipoaspirates. METHODS Human lipoaspirate (n = 8) was harvested using a standard liposuction technique. Stromal vascular fraction cells were separated from adipocytes and cultured to obtain purified adipose-derived stem cells. A panel of stem cell markers was used to identify the surface phenotypes of cultured adipose-derived stem cells. Three distinct stem cell subpopulations (CD90/CD45, CD105/CD45, and CD34/CD31) were selected from the stromal vascular fraction. Apoptosis and necrosis were determined by annexin V/propidium iodide assay and analyzed by flow cytometry. RESULTS The cultured adipose-derived stem cells demonstrated long-term proliferation and differentiation evidenced by cell doubling time and positive staining with oil red O and alkaline phosphatase. Isolated from lipoaspirates, adipocytes exhibited 19.7 ± 3.7 percent apoptosis and 1.1 ± 0.3 percent necrosis; stromal vascular fraction cells revealed 22.0 ± 6.3 percent of apoptosis and 11.2 ± 1.9 percent of necrosis; stromal vascular fraction cells had a higher rate of necrosis than adipocytes (p < 0.05). Among the stromal vascular fraction cells, 51.1 ± 3.7 percent expressed CD90/CD45, 7.5 ± 1.0 percent expressed CD105/CD45, and 26.4 ± 3.8 percent expressed CD34/CD31. CD34/CD31 adipose-derived stem cells had lower rates of apoptosis and necrosis compared with CD105/CD45 adipose-derived stem cells (p < 0.05). CONCLUSIONS Adipose-derived stem cells had a higher rate of apoptosis and necrosis than adipocytes. However, the extent of apoptosis and necrosis was significantly different among adipose-derived stem cell subpopulations.
Collapse
|
27
|
Abstract
In 2001, researchers at the University of California, Los Angeles, described the isolation of a new population of adult stem cells from liposuctioned adipose tissue. These stem cells, now known as adipose-derived stem cells or ADSCs, have gone on to become one of the most popular adult stem cells populations in the fields of stem cell research and regenerative medicine. As of today, thousands of research and clinical articles have been published using ASCs, describing their possible pluripotency in vitro, their uses in regenerative animal models, and their application to the clinic. This paper outlines the progress made in the ASC field since their initial description in 2001, describing their mesodermal, ectodermal, and endodermal potentials both in vitro and in vivo, their use in mediating inflammation and vascularization during tissue regeneration, and their potential for reprogramming into induced pluripotent cells.
Collapse
|
28
|
Hong SJ, Jia SX, Xie P, Xu W, Leung KP, Mustoe TA, Galiano RD. Topically delivered adipose derived stem cells show an activated-fibroblast phenotype and enhance granulation tissue formation in skin wounds. PLoS One 2013; 8:e55640. [PMID: 23383253 PMCID: PMC3561304 DOI: 10.1371/journal.pone.0055640] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/28/2012] [Indexed: 12/14/2022] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) are found in various tissues and can proliferate extensively in vitro. MSCs have been used in preclinical animal studies and clinical trials in many fields. Adipose derived stem cells (ASCs) have several advantages compared to other MSCs for use in cell-based treatments because they are easy to isolate with relative abundance. However, quantitative approaches for wound repair using ASCs have been limited because of lack of animal models which allow for quantification. Here, we addressed the effect of topically delivered ASCs in wound repair by quantitative analysis using the rabbit ear model. We characterized rabbit ASCs, and analyzed their multipotency in comparison to bone marrow derived-MSCs (BM-MSCs) and dermal fibroblasts (DFs) in vitro. Topically delivered ASCs increased granulation tissue formation in wounds when compared to saline controls, whereas BM-MSCs or DFs did not. These studies suggest that ASCs and BM-MSCs are not identical, though they have similar surface markers. We found that topically delivered ASCs are engrafted and proliferate in the wounds. We showed that transplanted ASCs exhibited activated fibroblast phenotype, increased endothelial cell recruitment, and enhanced macrophage recruitment in vivo.
Collapse
Affiliation(s)
- Seok Jong Hong
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
- * E-mail: (SJH); (RG)
| | - Sheng-Xian Jia
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
| | - Ping Xie
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
| | - Wei Xu
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
| | - Kai P. Leung
- Microbiology Branch, US Army Dental and Trauma Research Detachment, Institute of Surgical Research, Fort Sam Houston, Texas, United States of America
| | - Thomas A. Mustoe
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
| | - Robert D. Galiano
- Department of Surgery/Division of Plastic and Reconstructive Surgery, Laboratory for Wound Repair and Regenerative Medicine, Feinberg School of Medicine,Northwestern University, Chicago, Illinois, United States of America
- * E-mail: (SJH); (RG)
| |
Collapse
|
29
|
Efficacious and clinically relevant conditioned medium of human adipose-derived stem cells for therapeutic angiogenesis. Mol Ther 2013; 22:862-72. [PMID: 24413377 DOI: 10.1038/mt.2013.301] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 12/23/2013] [Indexed: 02/08/2023] Open
Abstract
Using stem cell-conditioned medium (CM) might be a viable alternative to stem cell transplantation, which is often hampered by low grafting efficiency and potential tumorigenesis, but the concentrations of angiogenic growth factors in CM are too low for therapeutic use and some components of the medium are not for human use. We used three-dimensional (3D) spheroid culture of human adipose-derived stem cells (ADSCs) with clinically relevant medium composed of amino acids, vitamins, glucose, and human serum to produce clinically relevant CM containing angiogenic and/or antiapoptotic factors such as vascular endothelial cell growth factor, fibroblast growth factor 2, hepatocyte growth factor, and chemokine (C-X-C motif) ligand 12. The concentrations of these factors were 23- to 27-fold higher than that in CM produced by conventional monolayer culture. Compared with injection of either monolayer culture CM or human ADSC, injection of spheroid culture CM to an ischemic region in mice significantly enhanced endothelial cell growth, CD34(+)/PTPRC(-) (endothelial progenitor) cell mobilization from bone marrow, and bone marrow cell homing to the ischemic region, resulting in improved blood vessel density, limb salvage, and blood perfusion in a mouse hindlimb ischemia model. The stem cell CM developed in this study will likely be an effective alternative to conventional stem cell transplantation therapy.
Collapse
|
30
|
Gimble JM, Bunnell BA, Guilak F. Human adipose-derived cells: an update on the transition to clinical translation. Regen Med 2012; 7:225-35. [PMID: 22397611 DOI: 10.2217/rme.11.119] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pace of discovery involving adipose-derived cells continues to accelerate at both the preclinical and clinical translational levels. Adipose tissue is a source of freshly isolated, heterogeneous stromal vascular fraction cells and culture-expanded, adherent and relatively homogeneous adipose stromal/stem cells. Both populations display regenerative capacity in soft and hard tissue repair, ischemic insults and autoimmune diseases. While their major mechanism of action has been attributed to both direct lineage differentiation and/or paracrine factor release, current evidence favors a paracrine mechanism. Over 40 clinical trials using adipose-derived cells conducted in 15 countries have been registered with the NIH, the majority of which are Phase I or Phase I/II safety studies. This review focuses on the literature of the past 2 years in order to assess the status of clinical and preclinical studies on adipose-derived cell therapies for regenerative medicine.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Center for Stem Cell Research & Regenerative Medicine, Department of Pharmacology, Tulane University Health Science Center, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
31
|
Quijada P, Toko H, Fischer KM, Bailey B, Reilly P, Hunt KD, Gude NA, Avitabile D, Sussman MA. Preservation of myocardial structure is enhanced by pim-1 engineering of bone marrow cells. Circ Res 2012; 111:77-86. [PMID: 22619278 DOI: 10.1161/circresaha.112.265207] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Bone marrow-derived cells to treat myocardial injury improve cardiac function and support beneficial cardiac remodeling. However, survival of stem cells is limited due to low proliferation of transferred cells. OBJECTIVE To demonstrate long-term potential of c-kit(+) bone marrow stem cells (BMCs) enhanced with Pim-1 kinase to promote positive cardiac remodeling. METHODS AND RESULTS Lentiviral modification of c-kit(+) BMCs to express Pim-1 (BMCeP) increases proliferation and expression of prosurvival proteins relative to BMCs expressing green fluorescent protein (BMCe). Intramyocardial delivery of BMCeP at time of infarction supports improvements in anterior wall dimensions and prevents left ventricle dilation compared with hearts treated with vehicle alone. Reduction of the akinetic left ventricular wall was observed in BMCeP-treated hearts at 4 and 12 weeks after infarction. Early recovery of cardiac function in BMCeP-injected hearts facilitated modest improvements in hemodynamic function up to 12 weeks after infarction between cell-treated groups. Persistence of BMCeP is improved relative to BMCe within the infarct together with increased recruitment of endogenous c-kit(+) cells. Delivery of BMC populations promotes cellular hypertrophy in the border and infarcted regions coupled with an upregulation of hypertrophic genes. Thus, BMCeP treatment yields improved structural remodeling of infarcted myocardium compared with control BMCs. CONCLUSIONS Genetic modification of BMCs with Pim-1 may serve as a therapeutic approach to promote recovery of myocardial structure. Future approaches may take advantage of salutary BMC actions in conjunction with other stem cell types to increase efficacy of cellular therapy and improve myocardial performance in the injured myocardium.
Collapse
|
32
|
Qayyum AA, Haack-Sørensen M, Mathiasen AB, Jørgensen E, Ekblond A, Kastrup J. Adipose-derived mesenchymal stromal cells for chronic myocardial ischemia (MyStromalCell Trial): study design. Regen Med 2012; 7:421-8. [DOI: 10.2217/rme.12.17] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue represents an abundant, accessible source of multipotent adipose-derived stromal cells (ADSCs). Animal studies have suggested that ADSCs have the potential to differentiate in vivo into endothelial cells and cardiomyocytes. This makes ADSCs a promising new cell source for regenerative therapy to replace injured tissue by creating new blood vessels and cardiomyocytes in patients with chronic ischemic heart disease. The aim of this special report is to review the present preclinical data leading to clinical stem cell therapy using ADSCs in patients with ischemic heart disease. In addition, we give an introduction to the first-in-man clinical trial, MyStromalCell Trial, which is a prospective, randomized, double-blind, placebo-controlled study using culture-expanded ADSCs obtained from adipose-derived cells from abdominal adipose tissue and stimulated with VEGF-A165 the week before treatment.
Collapse
Affiliation(s)
- Abbas Ali Qayyum
- Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| | - Mandana Haack-Sørensen
- Cardiology Stem Cell Laboratory, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| | - Anders Bruun Mathiasen
- Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| | - Erik Jørgensen
- Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| | - Annette Ekblond
- Cardiology Stem Cell Laboratory, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| | - Jens Kastrup
- Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, 9, Blegdamsvej, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
33
|
Cho SW, Sun HJ, Yang JY, Jung JY, Choi HJ, An JH, Kim SW, Kim SY, Park KJ, Shin CS. Human Adipose Tissue-Derived Stromal Cell Therapy Prevents Bone Loss in Ovariectomized Nude Mouse. Tissue Eng Part A 2012; 18:1067-78. [DOI: 10.1089/ten.tea.2011.0355] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Sun Wook Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jin Sun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Yeon Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Joo Yeon Jung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung Jin Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun An
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Yeon Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu-Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Prasongchean W, Ferretti P. Autologous stem cells for personalised medicine. N Biotechnol 2012; 29:641-50. [PMID: 22561284 DOI: 10.1016/j.nbt.2012.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 04/17/2012] [Accepted: 04/19/2012] [Indexed: 01/11/2023]
Abstract
Increasing understanding of stem cell biology, the ability to reprogramme differentiated cells to a pluripotent state and evidence of multipotency in certain adult somatic stem cells has opened the door to exciting therapeutic advances as well as a great deal of regulatory and ethical issues. Benefits will come from the possibility of modelling human diseases and develop individualised therapies, and from their use in transplantation and bioengineering. The use of autologous stem cells is highly desirable, as it avoids the problem of tissue rejection, and also reduces ethical and regulatory issues. Identification of the most appropriate cell sources for different potential applications, development of appropriate clinical grade methodologies and large scale well controlled clinical trials will be essential to assess safety and value of cell based therapies, which have been generating much hope, but are by and large not yet close to becoming standard clinical practice. We briefly discuss stem cells in the context of tissue repair and regenerative medicine, with a focus on individualised clinical approaches, and give examples of sources of autologous cells with potential for clinical intervention.
Collapse
|
35
|
Arsalan M, Woitek F, Adams V, Linke A, Barten MJ, Dhein S, Walther T, Mohr FW, Garbade J. Distribution of cardiac stem cells in the human heart. ISRN CARDIOLOGY 2012; 2012:483407. [PMID: 22462025 PMCID: PMC3312548 DOI: 10.5402/2012/483407] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/13/2011] [Indexed: 12/23/2022]
Abstract
Introduction.
The existence of human cardiac stem cells (hCSC) and their regenerative capacity are not fully defined. The aim of this study was to identify and analyse the distribution of hCSCs by flow cytometry (FCM). Methods. Tissue samples from the left ventricle (LV) and the appendages of the right atrium (RA) and left atrium (LA) were taken during cardiac surgery. Mononuclear cells (MNCs) were isolated, labelled for the stem-cell-marker c-kit and hematopoietic-lineage markers and analysed by FCM. Results. HCSCs could be isolated from the RA, LA, and LV without significant quantitative difference between both atria (A) (RA 4.80 ± 1.76% versus LA 4.99 ± 1.69% of isolated MNCs, P = 0.922). The number of hCSCs was significantly higher in both atria compared to the left ventricle (A 4.90 ± 1.29% versus LV 0.62 ± 0.14% of isolated MNCs, P = 0.035). Conclusion. The atria contain a higher concentration of hCSC than the left ventricle. HCSCs located in the atria could serve as an endogenous source for heart regeneration.
Collapse
Affiliation(s)
- Mani Arsalan
- Department of Cardiac Surgery, Kerckhoff Klinik, Bad Nauheim, Benekestr. 2-8, 61231 Bad Nauheim,, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mesenchymal stem cells and cardiovascular disease: a bench to bedside roadmap. Stem Cells Int 2012; 2012:175979. [PMID: 22315617 PMCID: PMC3270473 DOI: 10.1155/2012/175979] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/13/2011] [Indexed: 02/08/2023] Open
Abstract
In recent years, the incredible boost in stem cell research has kindled the expectations of both patients and physicians. Mesenchymal progenitors, owing to their availability, ease of manipulation, and therapeutic potential, have become one of the most attractive options for the treatment of a wide range of diseases, from cartilage defects to cardiac disorders. Moreover, their immunomodulatory capacity has opened up their allogenic use, consequently broadening the possibilities for their application. In this review, we will focus on their use in the therapy of myocardial infarction, looking at their characteristics, in vitro and in vivo mechanisms of action, as well as clinical trials.
Collapse
|