Abstract 1281: Human Adipose Tissue-derived Stem Cells Potently Preserve Cardiac Function Resulting From Permanently Induced Myocardial Ischemia

Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Liying Cai ◽  
Brian H Johnstone ◽  
Todd G Cook ◽  
Keith L March

Background The use of stem cells has gained much interest as a potential therapeutic approach for repair of damage caused by a variety of cardiac insults. We and others have demonstrated the ability of pluripotent adipose-derived stem cells (ASCs), to promote repair in ischemic skeletal tissues. Here we demonstrate that ASCs also stimulate a robust functional improvement following acute myocardial infarction (MI) in rats. Methods and Results ASCs were harvested from human subcutaneous adipose tissue. ASCs were characterized in vitro before in vivo testing. Growth and metabolic activity of human microvascular endothelial cells (HMVEC) cultured in growth-factor deficient minimal medium (MM) increased 1.7-fold when supplemented with a 1:1 mixture of ASC CM ( p <0.01). Sprout formation by HMVECs and migration of endothelial progenitor cell (EPC) was enhanced by 2.1 and 2.0-fold, repectively, when ASC CM was added to MM ( p <0.01). Following demonstration of potency in vitro, the ASCs were evaluated for the ability to protect and rescue ischemic myocardium in an athymic nude rat following permanent ligation of the proximal LAD region. Immediately after ligation 10 6 ASCs in 100 μl saline or carrier alone was injected into 2 sites of the peri-infarct region, then at 4 and 28 d heart function was evaluated echocardiography using a Visualsonics Vivo770. ASC-treated rats consistently exhibited better cardiac function at 1 month compared to the saline control group. LV ejection fraction of the ASC group was 56 ± 7% (mean ± SEM) vs 37 ± 3% for the control (p<0.04). Fractional shortening was 32 ± 5% (ASC) vs 19 ± 2% (p<0.04). LV volumes both at end-diastolic and end-systolic stages were lower in ASC group (311 ± 17 μl and 139 ± 21 μl, respectively) than saline group (391 ± 30 μl and 249 ± 27 μl) (p<0.03). Anterior wall thinning was attenuated in ASC group (1.6 ± .08 mm vs 1.2 ± .2, at end-diastole, p<0.03). Post-mortem histological analysis demonstrated that ASC treated hearts had lower fibrosis (26 ± 6% vs 34 ± 6%; p<0.05). Conclusion We have demonstrated that ASCs have great potential as a cell therapy to preserve heart function following ischemic insult. Given the abundant source of ASCs, therapies with these cells have a higher potential for widespread adoption compared to more rare cell types.

Gut ◽  
2008 ◽  
Vol 58 (4) ◽  
pp. 570-581 ◽  
Author(s):  
H Aurich ◽  
M Sgodda ◽  
P Kaltwasser ◽  
M Vetter ◽  
A Weise ◽  
...  

2019 ◽  
Vol 208 (3-4) ◽  
pp. 148-157
Author(s):  
Tao Luo ◽  
Xueqin Yang ◽  
Yan Sun ◽  
Xinqi Huang ◽  
Ling Zou ◽  
...  

Osteogenic differentiation of human adipose tissue-derived stem cells (hASCs) is a complex process that is regulated by multiple factors, including microRNAs (miRNAs). The miRNA miR-20a was shown to promote bone formation from bone marrow-derived mesenchymal stem cells. However, the role of miR-20a in osteogenic differentiation of hASCs remains unclear. In this study, we systematically evaluated the function of miR-20a in regulating hASC osteogenesis in vitro. hASCs were transduced with miR-20a-overexpressing and miR-20a-sponge lentiviral vectors, with green fluorescent protein (GFP) as a control. The results showed that miR-20a transcription was upregulated after hASC mineralization. Compared with the miR-20a-sponge, GFP, and hASC groups, the miR-20a-overexpressing group showed higher alkaline phosphatase (ALP) activity on days 7 and 14. Moreover, the mRNA level of ALP increased significantly in the miR-20a-overexpressing group on day 14. Furthermore, the protein of the target gene PPARγ was decreased, and the osteogenic differentiation-associated proteins ALP, osteocalcin, and RUNX2 were upregulated. hASCs anchored to HA/β-TCP revealed a healthy polygonal morphology and developed cytoplasmic extensions. miR-20a promoted osteogenic differentiation of the cell scaffold. Taken together, these data ­confirm that miRNA-20a promotes the osteogenesis of hASCs in vitro, and its essential role in vivo needs further ­investigation.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Xuemei Fan ◽  
Sheng He ◽  
Huifang Song ◽  
Wenjuan Yin ◽  
Jie Zhang ◽  
...  

Abstract Background The human endometrium in premenopausal women is an active site of physiological angiogenesis, with regenerative cells present, suggesting that the endometrium contains adult angiogenic stem cells. In the context of cardiac repair after ischemic injury, angiogenesis is a crucial process to rescue cardiomyocytes. We therefore investigated whether human endometrium-derived stem cells (hEMSCs) can be used for cardiac repair after ischemic injury and their possible underlying mechanisms. Methods Comparisons were made between hEMSCs successfully isolated from 22 premenopausal women and human bone marrow mesenchymal stem cells (hBMSCs) derived from 25 age-matched patients. Cell proliferation, migration, differentiation, and angiogenesis were evaluated through in vitro experiments, while the ability of hEMSCs to restore cardiac function was examined by in vivo cell transplantation into the infarcted nude rat hearts. Results In vitro data showed that hEMSCs had greater proliferative and migratory capacities, whereas hBMSCs had better adipogenic differentiation ability. Human umbilical cord vein endothelial cells, treated with conditioned medium from hEMSCs, had significantly higher tube formation than that from hBMSCs or control medium, indicating greater angiogenic potentials for hEMSCs. In vivo, hEMSC transplantation preserved cardiac function, decreased infarct size, and improved tissue repair post-injury. Cardiac metabolism, assessed by 18F-FDG uptake, showed that 18F-FDG uptake at the infarction area was significantly higher in both hBMSC and hEMSC groups, compared to the PBS control group, with hEMSCs having the highest uptake, suggesting hEMSC treatment improves cardiomyocyte metabolism and survival after injury. Mechanistic assessment of the angiogenic potential for hEMSCS revealed that angiogenesis-related factors angiopoietin 2, Fms-like tyrosine kinase 1, and FGF9 were significantly upregulated in hEMSC-implanted infarcted hearts, compared to the PBS control group. Conclusion hEMSCs, compared to hBMSCs, have greater capacity to induce angiogenesis, and improved cardiac function after ischemic injury.


Pharmaceutics ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 495
Author(s):  
Prakash Gangadaran ◽  
Ramya Lakshmi Rajendran ◽  
Ji Min Oh ◽  
Eun Jung Oh ◽  
Chae Moon Hong ◽  
...  

Angiogenesis is defined as the generation of new blood vessels or the sprouting of endothelial cells from a pre-existing vascular network. Angiogenesis occurs during the growth and development of an organism, the response of organs or tissues to injury, and during cancer development and progression. The majority of studies on stem-cell-derived extracellular vesicles (EVs) have used cell lines, and have primarily focused on well-known solitary proteins. Here, we isolated stem cells from human adipose tissue (ADSCs), and we isolated EVs from them (ADSC-EVs). The ADSC-EVs were characterised and 20 angiogenic proteins were analysed using an angiogenic antibody array. Furthermore, we analysed the ability of ADSC-EVs to induce angiogenesis in vitro and in vivo. ADSC-EVs were positive for CD81 and negative for GM130, calnexin, and cytochrome-C. ADSC-EVs showed typical EV spherical morphology and were ~200 nm in size. ADSC-EVs were found to contain angiogenic proteins as cargo, among which interleukin 8 (IL-8) was the most abundant, followed by chemokine (C-C motif) ligand 2 (CCL2), a tissue inhibitor of metalloproteinases 1 (TIMP-1), TIMP-2, and vascular endothelial growth factor-D (VEGF-D). ADSC-EVs treatment increased the proliferation, migration, total vessel length, total number of junctions, and junction density of endothelial cells in vitro. The results of an in vivo Matrigel plug assay revealed that ADSC-EVs induced more blood vessels in the Matrigel compared with the control. These results demonstrate that ADSC-EVs contain angiogenic proteins as cargo and promote angiogenesis in vitro and in vivo. Therefore, ADSC-EVs have potential for therapeutic use in ischaemia.


2007 ◽  
Vol 361 (2) ◽  
pp. 342-348 ◽  
Author(s):  
Christian Elabd ◽  
Chiara Chiellini ◽  
Ali Massoudi ◽  
Olivia Cochet ◽  
Laure-Emmanuelle Zaragosi ◽  
...  

2021 ◽  
Author(s):  
Xu Xu ◽  
Sujing Qiang ◽  
Lingyun Tao ◽  
Jie Zhou ◽  
Jing Ni

Abstract Background Mesenchymal stem cells (MSCs), kinds of seed cells, are expected to improve impaired diabetic cardiac function. Inflammation and autophagy play the important role in the development of metabolic disorder induced heart failure. The aim of this work was to assess the effect of adipose tissue derived mesenchymal stem cells (ADMSCs) on metabolic disorder induced heart failure and the underlying mechanisms. Methods In vivo, 8 weeks old male C57BL/6 mice were randomly divided into three groups: normal chaw mice (sham group), high fat diet fed and streptozotocin intraperitoneal injected mice (HFD + STZ group) and ADMSCs tail intravenous injected per week for 3 months after the mice were treated with HFD + STZ (ADMSCs + HFD + STZ group). The lipid and glucose levels as well as echocardiography were measured per week. Immunohistochemistry was used to detect the adhesion of macrophages in heart tissue among three groups. Besides, inflammatory cytokines such as interleukin-1β (IL-1β), tumor necrosis factor α (TNFα), interleukin-6 (IL-6) and interleukin-8(CXCL-15) were measured by western blot or RT-qPCR. In vitro, H9c2 cardiomyocytes were stimulated to 33mM glucose in the presence or absence of IL-1β. Transmission electron microscope, mRFG-GFP-LC3 assay and flow cytometry were used to investigate autophagy related apoptosis in H9c2 cells. Results HFD + STZ treated mice presented significant cardiac hypertrophy, body weight loss, hyperglycemia and hyperlipidemia. However, these changes were remarkably reversed by ADMSCs administration. The administration of ADMSCs also remit histological alterations and deposition of collagen in the heart tissue. Furthermore, ADMSCs downregulated the adhesion of macrophages in heart tissue. More importantly, IL-1β from macrophages increased the autophagy of myocardial cell stimulated with high glucose which eventually leaded to their apoptosis and the following cardiac dysfunction. Conclusions This study confirmed that ADMSCs may have potential for use in improving cardiac function by restraining autophagy and apoptosis of myocardial cell. We also found the roles of the IL-1β in hyperglycemia and hyperlipidemia induced cardiac injuries, which may be a key factor for diabetic complications.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3098-3098 ◽  
Author(s):  
Rosa M. Yañez ◽  
Maria L. Lamana ◽  
Javier Garcia-Castro ◽  
Manuel Ramirez ◽  
Juan A. Bueren

Abstract Previous studies have shown the immunomodulatory properties of bone marrow mesenchymal stem cells (BM-MSCs), opening the possibility of using these cells for the treatment of graft-versus-host disease (GVHD) in patients transplanted with allogeneic hematopoietic grafts. Additionally, Phase I studies in patients with Crohn’s disease suggested the efficacy of adipose tissue-derived mesenchymal stem cells (Ad-MSCs) for the healing of Crohn’s fistulas. In the present study we have investigated in vitro and in vivo, the immunomodulatory effects of Ad-MSCs, compared to BM-MSCs. We observed that both BM-MSCs and Ad-MSCs were negative for CD34, CD45, CD14, CD31 and MHC class I expression, while positive for CD29, CD44, CD90 and CD105. When studying the immunomodulatory effects of these cells in vitro, we found that - as happened with BM-MSCs - Ad-MSCs did not induce proliferation of allogeneic lymphocytes and were not lysed by cytotoxic T cells or alloreactive natural killer cells, indicating that Ad-MSCs are non-immunogenic. Additionally, the presence of Ad-MSCs inhibited in a dose-dependent fashion, both the mixed lymphocyte reaction (MLR) and the T cell proliferation induced by mitogens. To determine whether cell-to-cell contact between Ad-MSCs and PBMNCs was required for immunosuppression, transwell experiments were conducted. Phytohaemagglutinin (PHA)-stimulated lymphocytes were cultured in the upper chamber of a transwell, while irradiated Ad-MSCs remained in the lower chamber. As observed with BM-MSCs, Ad-MSCs were also capable of suppressing the lymphocytes proliferation in this transwell assay. When conditioned medium from Ad-MSCs was added to the MLR, the immuno-suppressive effect persisted, although at a lower level than that observed in a cell-to-cell contact system. Next we studied whether our in vitro findings were of significance in an in vivo mouse model of haploidentical transplantation. In these experiments irradiated F1(C57Bl/Balbc) recipient mice received 1x107 bone marrow cells from C57Bl mice, together with 2x107 splenocytes from the donor, to induce GVHD. One cohort of recipient mice received additional i.v. infusions of 5x105 mouse Ad-MSCs, administered at periodic intervals for up to 28 days post-transplant. When compared to the control group, the severity of the GVHD was significantly reduced in mice receiving Ad-MSCs. Our results suggest that Ad-MSCs obtained from adipose tissue may constitute a new and readily available source of immunomodulatory cells for the prophylaxis and/or treatment of GVHD in patients transplanted with allogeneic grafts.


Sign in / Sign up

Export Citation Format

Share Document